127
Research Collection Doctoral Thesis Identification of molecular mechanisms controlling lymphatic vessel formation by use of the embryonic stem cell-derived embryoid body assay Author(s): Marino, Daniela Publication Date: 2009 Permanent Link: https://doi.org/10.3929/ethz-a-006025517 Rights / License: In Copyright - Non-Commercial Use Permitted This page was generated automatically upon download from the ETH Zurich Research Collection . For more information please consult the Terms of use . ETH Library

Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

Research Collection

Doctoral Thesis

Identification of molecular mechanisms controlling lymphaticvessel formation by use of the embryonic stem cell-derivedembryoid body assay

Author(s): Marino, Daniela

Publication Date: 2009

Permanent Link: https://doi.org/10.3929/ethz-a-006025517

Rights / License: In Copyright - Non-Commercial Use Permitted

This page was generated automatically upon download from the ETH Zurich Research Collection. For moreinformation please consult the Terms of use.

ETH Library

Page 2: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

DISS. ETH Nr. 18687

Identification of molecular mechanisms controlling lymphatic vessel formation by use of the embryonic stem cell-derived embryoid body

assay

A dissertation submitted to ETH ZURICH

for the degree of Doctor of Sciences

presented by

Daniela Marino

Laurea Magistrale, Faculty of Biotechnology, University of Milan, Italy Born November 17, 1981

Citizen of Italy

Accepted on the recommendation of

Prof. Dr. Michael Detmar Prof. Dr. Cornelia Halin-Winter

2009

Page 3: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the
Page 4: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

"Fatti non foste a viver come bruti, ma per seguir virtute e canoscenza"

“You were not born to live like animals but to pursue virtue and possess knowledge”

(Dante Alighieri, Divina Commedia, Inferno canto XXVI, 116-120)

Page 5: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the
Page 6: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

Table of contents 1 SUMMARY 7

1.1 Summary 7 1.2 Riassunto 10

2 INTRODUCTION 13

2.1 The lymphatic vascular system: Embryonic development and role in health and disease 13

2.1.1 The lymphatic system history 16 2.1.2 Concepts of lymphatic vascular system development 17 2.1.3 Mechanisms of mammalian lymphatic vascular development 18 2.1.4 Lymphatic vascular development in Xenopus Laevis 28 2.1.5 Molecular markers of lymphatic endothelium 31 2.1.6 Molecular mediators of adult lymphangiogenesis 33 2.1.7 Involvement of lymphatic vessels in disease 35

2.2 Mouse embryonic stem cell-derived embryoid bodies 42

2.2.1 EB culture methods 44 2.2.2 Vasculogenesis, blood and lymphatic vascular development and (anti-) lymph-

angiogenesis research 45 3 AIM AND OUTLINE OF THIS DISSERTATION 48 4 IDENTIFICATION OF MOLECULAR MECHANISMS CONTROLLING LYMPHATIC

VESSEL FORMATION 49

4.1 A role for all-trans-retinoic acid in the early steps of lymphatic vasculature development 49

4.1.1 Abstract 49 4.1.2 Introduction 50 4.1.3 Materials and methods 52 4.1.4 Results 58 4.1.5 Discussion 73

4.2 Activation of the epidermal growth factor receptor promotes lymphangiogenesis in vitro

and in vivo 78

4.2.1 Abstract 78 4.2.2 Introduction 79 4.2.3 Materials and methods 80 4.2.4 Results 86 4.2.5 Discussion 98

5 CONCLUSIONS AND OUTLOOK 103 6 ACKNOWLEDGMENTS 106 7 REFERENCES 108 8 CURRICULUM VITAE 124

Page 7: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the
Page 8: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

7

1 SUMMARY

1.1 Summary

The lymphatic vascular system, the second vascular system present in vertebrates, has emerged

as a fundamental player in physiological, as well as pathological, processes. It maintains tissue

fluid balance, contributes to immune cell trafficking and absorbs lipids in the intestine. Recent

scientific findings have defined its role in lymphedema, obesity, inflammation, tumor metastasis

and hypertension. Considerable progress has been made in understanding the major mechanisms

controlling the formation of the lymphatic vascular system during development as well as

lymphatic vessel remodeling (lymphangiogenesis) in the adult organism. Nevertheless, further

studies are needed to develop therapeutic strategies for the effective control of lymphatic vessel

growth or regression in pathological conditions.

During embryogenesis, the lymphatic endothelial cells arise from the anterior cardinal veins of

the embryo and form lymphatic vessels in a stepwise manner that includes acquisition of

lymphatic competence and commitment, budding and sprouting, lymph sac formation and

vessel maturation. Analyses of genetically modified animal models have characterized the later

phases of lymphatic development; however, the processes that regulate the earliest steps remain

unknown. The first part of the present work aimed at unravelling the molecular mechanisms

controlling the earliest steps of lymphatic competence and commitment. Using mouse

embryonic stem cell-derived embryoid bodies (EBs), an in vitro model that largely mimics

mouse embryogenesis, we identified all-trans-retinoic acid (RA) as a novel regulator of the

expression of the lymphatic vessel endothelial receptor (LYVE)-1 (lymphatic competence) and

the transcription factor Prox1 (lymphatic commitment) by venous endothelial cells. The studies

also revealed a synergistic effect of cAMP on the induction of LYVE-1/Prox1 expression by

Page 9: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

8

RA. The use of antagonist molecules and immunohistochemical stainings indicated that retinoic

acid receptor (RAR)-α and cAMP-dependent protein kinase (Pk)-A are probable mediators of

the RA/cAMP activity in the lymphatic differentiation processes. Moreover, in vivo studies

revealed that exposure of mouse and Xenopus embryos to RA upregulated LYVE-1 and Prox1

expression in the endothelial cells of the cardinal veins and the developing primary lymph sacs.

In contrast, in utero exposure of mouse embryos to the RAR-α antagonist, Ro 41-5253,

decreased LYVE-1 and Prox1 expression in the same vascular regions. Taken together, these

findings indicate that RA regulates the earliest steps of lymphatic vasculature development.

Interestingly, intraperitoneal injection of RA in adult mice resulted in increased dermal

lymphatic vessel growth; thus, investigating the role of RA in adult lymphangiogenesis might

lead to the development of novel therapeutic strategies for e.g. lymphedema.

The second part of this thesis aimed at using the EB model to identify novel lymphangiogenic

pathways. To this end, EBs were treated with small organic molecules with well-defined

pathway specificities and the formation of lymphatic vessels was evaluated. The screening

identified the small molecule GW2974 (N4-(1-benzyl-1H-indazol-5-yl)-N6,N6-dimethyl-

pyrido[3,4-d]pyrimidine-4,6-diamine), which targets the tyrosine kinase associated with

epidermal growth factor receptor (EGFR/ErbB2), as an inhibitor of lymphatic vessel formation

in the EBs. Conversely, EBs treated with EGF, an activating ligand of EGFR/ErbB2, displayed

increased number of lymphatic vessel networks as compared to control EBs. We found that

human dermal lymphatic endothelial cells express low levels of EGFR; GW2974, as well as

EGFR/ErbB2 blocking antibodies, inhibited in vitro migration and tube formation of these cells

whereas EGF significantly induced those processes. In vivo, mice subcutaneously implanted

with matrigel plugs containing EGF displayed increased lymphatic vessel size and density in the

plug-associated skin. Moreover, analysis of skin from mice expressing amphiregulin (another

EGFR ligand) under control of the keratin 14 promoter revealed more and larger dermal

lymphatic vessels. These findings suggest that EGFR signalling has a role in lymphatic vessel

Page 10: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

9

formation and that targeting EGFR might represent a novel strategy to interfere with

lymphangiogenesis in pathological conditions, such as tumor metastasis.

Collectively, our results identify an important unexpected role for retinoic acid in lymphatic

vascular system development and a novel function for EGFR signalling in lymphangiogenesis.

Page 11: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

10

1.2 Riassunto

Il sistema linfatico, il secondo sistema vascolare presente nei vertebrati, si è rivelato essenziale

sia in processi fisiologici che patologici. Esso, infatti, mantiene l’equilibrio dei fluidi tissutali,

contribuisce al trasporto delle cellule immunitarie e assorbe i lipidi nell’intestino. Recenti

ricerche scientifiche, inoltre, hanno riconosciuto il ruolo di questo sistema vascolare in patologie

quali linfedema, obesità, infiammazione, metastasi tumorale ed ipertensione. Nonostante sia

stato raggiunto un progresso straordinario nella comprensione dei meccanismi principali che

controllano la formazione del sistema linfatico durante lo sviluppo embrionale così come la

crescita dei vasi linfatici nell’organismo adulto (linfangiogenesi), ulteriori studi sono necessari

per sviluppare strategie terapeutiche che possano efficacemente controllare la crescita o la

regressione dei vasi linfatici in situazioni patologiche.

In particolare, durante lo sviluppo embrionale, le cellule endoteliali linfatiche originano dalle

vene cardinali anteriori dell’embrione e formano i vasi linfatici in un processo a stadi che

include l’acquisizione della competenza e della specificazione linfatica, migrazione, formazione

dei sacchi linfatici e maturazione dei vasi. L’analisi di modelli animali geneticamente modificati

ha contribuito a caratterizzare le fasi finali dello sviluppo linfatico, d’altrocanto però, i processi

che regolano le fasi iniziali rimangono, a tutt’oggi, sconosciuti. La prima parte del presente

lavoro ha avuto come scopo l’identificazione dei processi molecolari che controllano

l’acquisizione della competenza e della specificazione linfatica. Mediante l’uso di corpi

embrionici ottenuti da cellule staminali embrionali di topo (EBs), modello in vitro, questo, che

ampiamente mima l’embriogenesi di topo in vivo, abbiamo scoperto come l’acido retinoico

(RA) regoli l’espressione del gene LYVE-1 (competenza linfatica) e del gene Prox1

(specificazione linfatica) nelle cellule endoteliali venose. Questi studi in vitro, hanno inoltre

evidenziato un effetto sinergistico di cAMP nell’ attivazione dell’espressione di LYVE-1/Prox1

Page 12: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

11

da parte di RA. L’utilizzo di molecole antagoniste e di colorazioni immunoistochimiche hanno

suggerito che il recettore per l’acido retinoico (RAR)-α e la proteina chinasi dipendente da

cAMP (PkA) potrebbero essere i mediatori dell’attività di RA/cAMP nei processi di

differenziamento linfatico. Inoltre, studi in vivo, hanno rivelato che l’esposizione all’RA di

embrioni di topo e di rana (Xenopus laevis) ha indotto una maggiore espressione di LYVE-1 e

Prox1 nelle cellule endoteliali delle vene cardinali e dei nascenti sacchi linfatici. Al contrario,

l’esposizione in utero all’antagonista del RAR-α, Ro 41-5253, ha diminuito l’espressione di

LYVE-1 e Prox1 nelle stesse regioni vascolari. Riassumendo, i nostri risultati indicano come

RA regoli le fasi iniziali dello sviluppo del sistema vascolare linfatico. Inoltre, è interessante

notare come un’ iniezione intraperitoneale di RA in topi adulti, abbia accresciuto la formazione

dei vasi linfatici (risultati non mostrati); quindi, ulteriori studi sul ruolo di RA nella

linfangiogenesi adulta potrebbe contribuire allo sviluppo di nuove strategie terapeutiche per

poter, per esempio, curare il linfedema.

La seconda parte di questa tesi, ha avuto lo scopo di identificare nuovi pathway linfangiogenici,

mediate l’utilizzo dei modelli EB. Per questo, gli EBs sono stati prima trattati con piccole

molecole organiche aventi specificità per definiti pathway molecolari e poi analizzati in termini

di crescita di vasi linfatici. Lo screening, ha rivelato come la molecola GW2974 (N4-(1-benzyl-

1H-indazol-5-yl)-N6,N6-dimethyl-pyrido[3,4-d]pyrimidine-4,6-diamine), che ha come target la

tirosin chinasi associata al recettore del fattore di crescita epidermale (EGFR-ErbB2), agisca da

inibitore della formazione di vasi linfatici negli EBs. Al contrario, EBs trattati con EGF, un

ligando che attiva EGFR-ErbB2, hanno presentato un numero maggiore di reti linfatiche rispetto

agli EB non trattati. Inoltre, abbiamo scoperto che le cellule endoteliali linfatiche umane

esprimono bassi livelli di EGFR. GW2974, così come anticorpi che bloccano l’EGFR-ErbB2,

hanno inibito la migrazione e la formazione di vasi in vitro di queste cellule, mentre EGF le ha

indotte in modo significativo. In vivo, topi a cui è stato impiantato matrigel contenente EGF,

hanno mostrato un incremento di misura e densità dei vasi linfatici nella pelle in contatto con il

Page 13: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

12

gel. Inoltre, l’analisi di campioni di pelle prelevati da topi transgenici che overesprimono

l’anfiregulina (un altro ligando per EGFR-ErbB2) sotto il controllo del promotore del gene per

la keratina 14, ha rivelato più e più grandi vasi linfatici dermali. Queste scoperte suggeriscono

come l’EGFR-ErbB2 abbia un ruolo nella formazione dei vasi linfatici e come il targeting

dell’EGFR-ErbB2 possa rappresentare una nuova strategia per interferire con la linfangiogensi

in condizioni patologiche quali, ad esempio, la metastasi tumorale. Riassumento, i nostri

risultati identificano un importante inaspettato ruolo dell’acido retinoico nello sviluppo dei vasi

linfatici durante l’embriogenesi e una nuova funzione per l’EGFR-ErbB2 nella linfangiogenesi.

Page 14: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

13

2 INTRODUCTION

2.1 The lymphatic vascular system: Embryonic development

and role in health and disease

Apart from the cardiovascular system, vertebrates also possess a lymphatic system that consists

of lymphatic vessels and lymphoid organs (lymph nodes, spleen, thymus, tonsils, Peyer’s

patches and lymphoid tissues associated with the respiratory systems). The organization and

function of the two vascular systems are distinct. The cardiovascular system is a closed

circulatory system with a central pump, the heart that distributes blood cells, oxygen, nutrients,

and hormones to all tissues. In contrast, the lymphatic vasculature is a one-way system that

removes fluid, cells, macromolecules, microbes and other substances from interstitial spaces

without a central driving force.

The lymphatic vascular system consists of blind-ended capillaries that collect and transport

extravasated protein-rich fluid that originates from blood serum, the lymph, from tissues to

precollector lymphatic vessels (Sacchi et al. 1997) then to collecting lymphatic vessels that

converge into lymphatic trunks to finally return the lymph to the venous circulation via the

thoracic duct through the connection with jugular and subclavian veins (Casley-Smith 1980). In

addition, in the intestine, lacteal lymphatic vessels, which are found in the villi, absorb and

transport vitamins and dietary fat released in form of lipid particles called chylomicrons (Jurisic

and Detmar 2009). Furthermore, the lymphatic system carries antigens and antigen-presenting

cells from the interstitium to the lymph nodes where they are presented to B and T cells (Jurisic

and Detmar 2009). Recently, interest in basic lymphatic research has been enhanced by

evidence demonstrating that the lymphatic system is involved in a number of human

Page 15: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

14

pathological conditions, such as lymphedema, inflammation, cancer metastasis (Cueni and

Detmar 2008) and hypertension (Machnik et al. 2009). The lymphatic vasculature is present in

the skin and in several internal organs, with the exception of the epidermis, nails, hairs,

cartilage, cornea, brain, bone marrow and retina. Lymphatic capillaries are lined by a single

layer of overlapping lymphatic endothelial cells (LECs) and lack a continuous basement

membrane (Figure 2.1.1) as well as pericyte or smooth muscle cell coverage. LECs of the

capillaries are oak leaf shaped, inter-connected by button-like junctions and linked to the

extracellular matrix by anchoring elastic fibers (Leak and Burke 1968; Gerli et al. 1991; Baluk

et al. 2007) (Figure 2.1.1). Under high interstitial pressure conditions, the junctions open via

anchoring fibrillin filaments stretching, thus allowing fluid, macromolecules and cells to move

into the vessel lumen. Overlapping endothelial cell-cell contacts, or primary valves, prevent

fluid from escaping the capillaries (Trzewik et al. 2001; Schmid-Schonbein 2003). Lymphatic

capillaries then merge into pre-collector vessels (still presenting anchoring filaments in some

areas) that, together with downstream collecting lymphatic vessels, present zipper-like

junctions, a basement membrane and smooth muscle cells (SMCs) (Scavelli et al. 2004). The

contractile activity of the SMCs and of the skeletal muscles, together with the respiratory

movement and the presence of intraluminal valves, promote lymph flow (Von der Weid and

Zawieja 2004).

Page 16: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

15

Figure 2.1.1 Structure of lymphatic vessels (Karpanen and Alitalo 2008). In contrast to blood vessels,

lymphatic vessels have thin walls and a relatively wide lumen. The endothelial cells of lymphatic capillaries

(green) partly overlap, forming valve-like openings, which allow easy access for fluid, macromolecules, and cells

into the vessel lumen. Lymphatic capillaries lack smooth muscle cells (SMCs; red) and have no basement

membrane. Anchoring filaments connect lymphatic capillary endothelial cells to the surrounding extracellular

matrix. The lymph drains from the lymphatic capillaries to precollecting and collecting lymphatic vessels, which

are finally emptied into veins in the jugular region. The precollecting and collecting lymphatic vessels have a

basement membrane, are surrounded by vascular smooth muscle cells and contain valves that prevent backflow of

the lymph. On its way, the lymph is filtered through a series of lymph nodes. In contrast, the endothelial cells of

blood vessels have a distinct basement membrane and are surrounded by pericytes/vSMCs, which form one or

multiple layers increasing in thickness with vessel size.

Page 17: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

16

2.1.1 The lymphatic system history

Scientific advances in the 17th century were the result of a new concept of science that coupled

experimentation and innovation. However, in the case of human anatomy, physicians and

scientists were limited to observation alone. Most of the descriptions and theories of human

anatomy were established by the Greek physician Galen in the second century. Galen developed

theories on the physiology of human blood circulation, but the role of the lymphatic system was

not discussed despite the fact that earlier the Greek anatomist Erasistratus (ca. 304-ca.250

B.C.E.) had described the presence of vessels containing “white blood” in human cadavers

(Lord 1968; Vegetti 1995). Finally, on 23rd July 1622, the Italian physician Gaspare Aselli

(1581-1626), while studying the digestive system of a dog, noted that some glands and

capillaries contained a cloudy fluid. In his book “De lactibus sive lacteis venis” ((Asellius 1627)

the first anatomical report in color, Figure 2.1.2), Aselli incorrectly concluded that these glands

were associated with the blood circulation and that the content was transported by “milky veins”

to the liver (at that time, still considered the center of the venous system).

Figure 2.1.2 De lactibus sive lacteis venis. In 1622, the Italian physician Gaspare Aselli, while studying the

digestive tract of a dog (A), noted glands and capillaries containing a milky fluid (white lines). He reported his

findings in the De lactibus sive lacteis venis (1627, B): the first anatomical report in color (C).

Page 18: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

17

Aselli’s observation inspired other scientists, like the French Jean Pecquet (1622-1674) who, in

a dog, found a lumbar reservoir draining the lacteals, now known as the cysterna chyli. The

Danish physician Thomas Bartholin (1616-1680) was the first to describe the lymphatic system

in humans (Eales 1974); in 1652 he published the “De Lacteis Thoracsis” where he described

the anatomy of the thoracic duct and recognised the lacteal veins as a second vascular system

within the body that he named the lymphatic system. Years later William Hunter (1718-1713)

and Alexander Monro (1733-1817) described the absorbent nature of the lymphatic vessels

draining fluid from the tissue and bringing it back to the blood circulation (Eales 1974).

Although the description of the anatomy of the lymphatic vascular system was progressing

rapidly, its embryonic origin remained unclear for a long time until Florence R. Sabin (1902-

1904) and F. Lewis (1905) postulated the “centrifugal theory” proposing that LECs derive from

the embryonic venous endothelium (Sabin 1902). An alternative “centripetal theory” postulated

by Huntington and Mc Clure in 1910 suggested that LECs arise from mesenchymal progenitor

cells, a lymphangioblast (Huntington and McClure 1910). These two opposing theories have

been the center of a century-old debate that nowadays seems to have been partially resolved.

2.1.2 Concepts of lymphatic vascular system development

The centrifugal theory proposes that in the developing embryo endothelial cells bud out from

the anterior cardinal veins, proliferate, migrate and form primary jugular lymph sacs (Oliver

2004). From the sacs, the endothelial cells sprout and form vessel networks throughout the

body. Several recent studies, using genetically engineered mouse models (Wigle et al. 2002),

lineage tracing experiments (Srinivasan et al. 2007) and in vivo imaging studies in zebrafish

(Yaniv et al. 2006) have demonstrated that lymphatic endothelial cells of venous origin are the

main source for the developing lymphatic vasculature. However, a dual origin from both

embryonic veins and from a mesenchymal ancestor has also been proposed. Studies in chicken

Page 19: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

18

embryos, suggest that the superficial parts of the jugular lymph sacs, as well as dermal

lymphatics, are derived from local lymphangioblasts (Wilting et al. 2006). In Xenopus tadpoles,

mesodermal precursor cells contribute to lymphatic vessel formation (Ny et al. 2005).

Furthermore, scattered mesenchymal cells, co-expressing lymphatic and leukocyte markers

were detected in mouse embryos in the jugular regions (Buttler et al. 2006), and cells co-

expressing lymphatic and macrophage markers have been suggested to integrate into lymphatic

vessels (Buttler et al. 2008).

2.1.3 Mechanisms of mammalian lymphatic vascular development

2.1.3.1 Lymphatic competence

In the mouse embryo, the lymphatic vascular system begins to form after the establishment of

the intra-embryonic blood vasculature (Figure 2.1.3) In particular, at embryonic day (ED) 9,

endothelial cells lining the anterior cardinal veins start to express lymphatic endothelial

hyaluronan receptor-1 (LYVE-1) indicating lymphatic competence, the first step of lymphatic

differentiation. It has been proposed that the acquisition of competence, prepares the endothelial

cells to receive further lymphatic specification signals (Wigle et al. 2002). LYVE-1, an

homologue of the blood vascular endothelium-specific hyaluronan receptor CD44 (Banerji et al.

1999), is a member of the link protein superfamily and was identified as a cell surface protein of

lymphatic endothelial cells (Banerji et al. 1999; Jackson et al. 2001; Prevo et al. 2001). The

molecular mechanisms that control LYVE-1 expression have not been identified; however, a

recent study has shown that the chicken ovalbumin upstream promoter-transcription factor

(COUP-TF)-II regulates its expression in human cultured LECs (Lee et al. 2009). The function

of LYVE-1 is still unclear; LYVE-1 knockout mice do not display any lymphatic vascular

Page 20: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

19

defects (Gale et al. 2007). However, mice deficient for the cell surface retention binding

protein-1 (CRSBP-1), a gene that is identical to LYVE-1, exhibit identifiable morphological and

functional alterations of lymphatic capillary vessels in certain tissues, such as the intestine,

marked by the constitutively increased interstitial-lymphatic flow and lack of typical irregularly-

shaped lumens (Huang et al. 2006). In the adult, LYVE-1 expression is maintained by the LECs

and it is one of the most used markers for lymphatic vessels. Its expression is also found in a

subset of macrophages (Maruyama et al. 2005; Schledzewski et al. 2006) and in liver sinusoids

(Mouta Carreira et al. 2001; Nonaka et al. 2007).

2.1.3.2 Lymphatic commitment

At ED 9.5, polarized expression of the transcription factor prospero-related homeobox (Prox)-1

is detected on one side of the anterior cardinal veins, thus conferring lymphatic commitment to

the LYVE-1+, lymphatic-competent endothelial cells (Figure 2.1.3). Prox1 is the most specific

lineage marker for lymphatic endothelium (Oliver and Detmar 2002) and it controls the initial

steps of lymphatic development. Inactivation of Prox1 in mice results in complete absence of

lymphatic vascular structures (Wigle et al. 2002) and in utero death at around ED 14.5. In the

absence of Prox1, endothelial cells that have budded from the cardinal veins continue to express

blood vascular endothelial cell (BECs) lineage-specific genes. Conversely, ectopic expression of

Prox1 in human BECs in vitro induces expression of lymphatic vascular signature genes (Hong

et al. 2002; Petrova et al. 2002; Wigle et al. 2002), such as VEGFR-3 (Petrova et al. 2002),

integrin α9 (Mishima et al. 2007) and FGFR-3 (Shin et al. 2006). siRNA knock down of Prox1

in LECs downregulates VEGFR-3, podoplanin and FGFR-3 and upregulates the BEC signature

genes neuropilin-1, ICAM (inter-cellular adhesion molecule 1) and MCP1 (monocyte

chemotactic protein-1) (Lee et al. 2009). At present the signals that induce Prox1 expression in a

restricted subpopulation of the embryonic cardinal veins are not well understood. Recently, it

Page 21: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

20

has been shown that vessels in mice deficient of SOX18, a transcription factor that belongs to

the SRY-relates HMG domain family, do not undergo definitive lymphatic differentiation.

Furthermore, SOX18 is expressed by the endothelial cells lining the dorsolateral sector of the

cardinal veins as early as ED 9, before Prox1 expression has begun. SOX18 expression remains

in the budding and jugular lymph sac forming lymphatic progenitor cells but it is not detectable

during lymphatic vascular system maturation or in the adult lymphatic vessels. These findings

suggest that SOX18 is not required for maintenance of LECs once they are fully differentiated.

Studies on the Prox1 promoter show that SOX18 directly activates this lymphatic master gene

(Francois et al. 2008). In addition, COUP-TF II physically interacts with Prox1 to form a stable

complex in various cell types including LECs (Lee et al. 2009). COUP-TF II is thought to

function as a coregulator of Prox1 and to be required, along with Prox1, to maintain the LEC

phenotype. Interestingly, COUP-TF II expression has been found on CD31+/Prox1+/LYVE-1+

endothelial cells of embryonic cardinal veins and on dermal adult lymphatic vessels in vitro and

in vivo (Lee et al. 2009). Mice lacking COUP-TF II die in utero around ED 10 and present head

and heart defects, hemorrhages, edema, enlarged blood vessels and malformed cardinal veins; it

still remains to be elucidated whether the presence of COUP-TF II is essential for lymphatic

differentiation.

2.1.3.3 Budding, migration and proliferation of LECs

Starting from ED 10.5, lymphatic committed embryonic endothelial cells response to vascular

endothelial growth factor (VEGF)-C, a ligand for VEGFR-3 and start to bud out from the

cardinal veins, proliferate, migrate and form the primary jugular lymph sacs (Tammela et al.

2005; Karpanen and Alitalo 2008) (Figure 2.1.3). During embryonic development, VEGFR-3 is

expressed by all endothelial cells and it is important for blood vessel remodeling. Indeed,

VEGFR-3 null mice die in utero at ED 9.5 due to severe cardiovascular defects (Dumont et al.

Page 22: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

21

1998; Hamada et al. 2000) while VEGF-C null mice do not develop a lymphatic vasculature and

die in utero at ED 17-19 (Karkkainen et al. 2004). VEGF-C is expressed by mesenchymal cells

in the jugular area of developing embryos. In its absence, the committed Prox1+ endothelial

cells arise normally in the cardinal veins but fail to sprout and migrate. VEGFR-3 has another

known ligand, VEGF-D; studies in VEGF-D deficient mice have demonstrated that this protein

is not essential for lymphatic vasculature development (Karkkainen et al. 2004; Baldwin et al.

2005). Recently, integrins have been shown to be involved in lymphatic vascular system

development; mice deficient for integrin α9 β1 die soon after birth because of chylothorax

(Huang et al. 2000). Integrin α9 β1 is a receptor for VEGF-C and D (Vlahakis et al. 2005) and

its stimulation, by extracellular matrix (ECM) components, induces tyrosine phosphorylation of

VEGFR-3 (Wang et al. 2001). Since VEGF-C and -D null mice, unlike to the VEGFR-3 null,

have no blood vascular phenotype it is thought that integrins contribute to VEGFR-3 signalling

in blood vessel remodeling (Haiko et al. 2008). Furthermore, VEGFR-3 interacts with

neuropilin-2 (NRP2) (Karpanen et al. 2006) and mice lacking NRP2, a receptor for class III

semaphorins, have abnormal lymphatic capillaries, while large vessel develop normally (Yuan

et al. 2002).

2.1.3.4 Sprouting from the lymphatic plexus

Starting from ED 11.5, the LECs that formed the jugular lymph sac proliferate and sprout to

expand the primary lymphatic plexuses (Figure 2.1.3). Recently, adrenomedullin has been

shown to play a role in this process. Adrenomedullin is a multifunctional vasodilator peptide

that transduces its effects through the calcitonin receptor-like receptor (calcrl) when the receptor

is associated with a receptor activity-modifying protein (RAMP)-2. Adrenomedullin-, calcrl-, or

RAMP2-null mice die during mid-gestation after developing interstitial lymphedema. A calcrl

conditional knockout in endothelial cells resulted in abnormal jugular lymphatic vessels due to

Page 23: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

22

impaired lymphatic endothelial cell proliferation (Fritz-Six et al. 2008). In addition, the

transcription factor NFATc1 (nuclear factor of activated T-cells-1), which is critical for lineage

selection in T-cell differentiation, cardiac valve morphogenesis and osteoclastogenesis, has been

shown to also have a role in lymphatic development and patterning. NFAT is colocalized with

the lymphatic markers Prox1, VEGFR-3 and podoplanin on cardinal vein endothelial cells as

LECs are committed. In both NFATc1 null and in utero treated embryos with the calcineurin,

(which is required for NFAT activation) inhibitor cyclosporine-A, Prox1, VEGFR-3 and

podoplanin positive endothelial cells budded from the cardinal veins at E11.5, but poorly

sprouted off the jugular lymph sacs to form the lymphatic plexus. These results, thus, suggest a

role for calcineurin-NFAT signalling in embryonic lymphatic patterning (Kulkarni et al. 2009).

2.1.3.5 Separation of blood and lymphatic vessels

Later in development (ED13.5-14.5), the first lymphatic vessels completely separate from the

blood vasculature (Figure 2.1.3). The tyrosine kinase, Syk, and the adaptor protein, SLP76, that

are almost exclusively expressed by hematopoietic cells are fundamental for this separation

process. Mice lacking Syk/Slp76 die perinatally due to haemorrhaging (Abtahian et al. 2003;

Sebzda et al. 2006). Recently, phospolipase gamma (PLCγ)-2, which is a downstream target of

Syk/SLP76, has been shown to play an essential role in initiating and maintaining the separation

of the blood and lymphatic vasculature (Ichise et al. 2009). Podoplanin, a small transmembrane

mucin-like protein, is highly expressed in lymphatic endothelial cells starting at ED 11.

Podoplanin null mice die at birth because of abnormal lung development and display impaired

lymphatic transport, dilation of lymphatic vessels and congenital lymphedema (Schacht et al.

2003). The precise molecular function of podoplanin has not been yet identified. However,

podoplanin is known to be involved in cytoskeletal organization of endothelial and tumor cells

(Schacht et al. 2003; Martin-Villar et al. 2006; Wicki et al. 2006) and to aggregate platelets via

Page 24: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

23

interaction with the C-type lectin-like receptor (CLEC)-2 (Kato et al. 2003; Suzuki-Inoue et al.

2007) which signals via Syk and SLP76 (Suzuki-Inoue et al. 2007). Finally, a recent study

reported blood-filled lymphatic vessel phenotype in mice lacking, or with reduced, podoplanin

expression (Fu et al. 2008). Thus, given the phenotypes of Syk/SLP76, PLCγ2 and podoplanin

null mice one could speculate on a possible function for podoplanin in forming thrombi to

prevent anastomoses between blood and lymphatic vessels. In addition, the phenotype of mice

null for sprouty-related ena/VASP homology domain-containing proteins (Spred) 1/2

(Taniguchi et al. 2007), two negative regulators of growth factor and ERK activation, resembles

that of Syk/SLP76 knockouts. Furthermore, studies on angiopoietin-like (Angptl)-4, have

suggested that organ-specific mechanisms are essential after birth to maintain blood and

lymphatic vessels separated. In fact, mice deficient for Angptl-4, display blood filled intestinal

lymphatics (Backhed et al. 2007).

2.1.3.6 Maturation of lymphatic vessel network

Primary lymphatic vessels undergo drastic changes to form a complex and organized network of

lymphatic capillaries and collectors (Figure 2.1.3). Foxc2 is a forkhead transcription factor that

is highly expressed in the developing lymphatic vasculature. Initial lymphatic vascular

development proceeds normally in absence of Foxc2. However, later on, collecting lymphatics

fail to develop valves, and capillaries acquire an excessive smooth muscle cell (SMC) coverage.

A similar phenotype has also been reported for mice lacking the PDZ domain of ephrinB2

(Makinen et al. 2005). Recently, it has been shown that Foxc2 controls lymphatic vessel

maturation through cooperation with NFATc1 (Norrmen et al. 2009). Angiopoietin (Ang)-2 is

also required for the proper development of the lymphatic vasculature. Ang2 -/- mice display

lymphatic vessel disorganization and hyperplasia, impaired recruitment of SMCs into collecting

lymphatic vessels, as well as irregularly patterned lymphatic capillaries (Gale et al. 2002).

Page 25: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

24

Importantly, studies on integrins and on elastic microfibril-associated proteins such as emilin-1,

(elastin microfibril interfacer-1) revealed an importance for interactions between the LECs and

the ECM. Mice lacking integrin α9 β1 develop respiratory failure, caused by an accumulation of

large volumes of pleural fluid, and die between 6 and 12 days of age (Huang et al. 2000). In

emilin-1-/- mice lymphatic drainage is impaired due to disorganized lymphatic vessels with

reduced anchoring filaments (Danussi et al. 2008). A similar phenotype to the one observed in

integrin α9 β1 -/- mice, is reported also for Net, a transcription factor member of the ternary

complex factor family which is expressed in sites of vasculogenesis during mouse development

(Ayadi et al. 2001). Moreover, Aspp1, the apoptosis stimulating protein of p53, was recently,

found to be involved in lymphatic vessel assembly as mice lacking Aspp1 have a disorganized

lymphatic plexus and impaired lymphatic drainage in the embryonic skin. Interestingly, the

drainage defects are resolved in the adult animals (Hirashima et al. 2008). Furthermore, targeted

inactivation of neuropilin (Nrp)-2 results in absence or severe reduction of small lymphatic

vessels and capillaries during development (Yuan et al. 2002). Finally, mice generated with

mutations in the phospoinositide 3-kinase (pi3kca) gene display defective development of the

lymphatic vasculature, resulting in perinatal appearance of chylous ascites and reduced number

of lymphatic capillaries (Gupta et al. 2007).

All known factors involved in lymphatic vascular system development are listed in Table 2.1.1

Page 26: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

25

Page 27: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

26

Table 2.1.1 Genetic mouse models displaying a lymphatic phenotype

Genes Function Model Lethality Lymphatic phenotype

Prox11 Transcription factor

KO E14.5 No lymphatic vasculature (-/-), adult-onset obesity, chylous ascites ( -/+ )

LYVE-12 Hyaluronan receptor

KO Normal None or only subtle defects: increased interstitial lymphatic flow, atypical shape of vessel lumen

VEGF-C3 Growth factor KO E17-19 No lymphatic vasculature (-/-), delayed lymphatic vascular development, lymphedema (-/+)

VEGFR-34 Growth factor receptor

KO E10 Cardiovascular failure, defective remodeling of vascular networks

Syk/SLP-76 5 Tyrosine kinase

KO Perinatal Abnormal blood-lymphatic connections, chylous ascites, defect in hematopoietic endothelial progenitors

Spred1/2 6 Neg. regulators of ERK

KO E12.5-15.5

Lymphedema, dilated and blood filled lymphatic vessels

Angpl4 7

Secreted inhibitor of lipoprotein lipase

KO < 3 weeksPostnatal lymphatic-venous partitioning defect in the small intestine

Podoplanin 8a Membrane glycoprotein

KO Perinatal

Lymphedema, dilatation and mispatterning of lymphatic vessels, diminished lymphatic transport. Blood filled lymphatic vessels 8b

Foxc2 9 Transcription factor

KO E12.5-Perinatal

Lymphatic hyperplasia, retrograde lymph flow ( -/+ ), abnormal patterning and pericyte investment of lymphatic vessels, absence of valves, lymphatic dysfunction (-/-)

EphrinB2 10 Ligand of EphB receptors

Mutant Perinatal Defective remodeling of lymphatic vascular network, hyperplasia, lack of valves, chylothorax

Neuropilin-2 11 Growth factor receptor

KO Postnatal (50%)

Transient absence or severe reduction of small lymphatic vessels and capillaries during development

Angiopoietin-2 12

Growth factor KO Perinatal Chylous ascites and subcutaneous edema, abnormal patterning of lymphatic vessels

Aspp1 13 p-53 binding KO Normal

Subcutaneous edema, disorganized and non-functional lymphatic vasculature in embryo, mispatterned collecting lymphatic vessels in adult

Page 28: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

27

Adrenomedullin 14

Vasoactive peptide

KO E14.5 Interstitial lymphedema (KO), abnormal jugular lymphatic vessels (conditional KO in ECs)

Emilin-1 15 ECM protein KO Normal Hyperplastic, enlarged, irregularly patterned lymphatic vessels, reduction of anchoring filaments, lymphedema

Integrinα9 16 Adhesion receptor

KO Perinatal Chylothorax, lymphedema

PLCγ2 17 Phospholipase Mutant Perinatal Abnormal blood-lymphatic connections, blood filled lymphatic vessels

NFATc1 18 Transcription factor

KO E12.5 Abnormal formation of jugular lymph sac

Net 19 Transcription factor

KO Perinatal Chylothorax, dilated lymphatic vessels

Sox18 20 Transcription factor

KO E14.5 Edema, chylous ascites, cardiovascular and hair follicle defects

Pi3kca 21 Phosphoinositide 3-kinase

MutantPostnatal (50%)

Chylous ascites, reduction of lymphatic capillaries

VEGF-C 22 Growth factor Tg/K14 Hyperplastic lymphatic vessels

VEGF-D 23 Growth factor Tg/K14 Hyperplastic lymphatic vessels

VEGF-A 24 Growth factor Tg/K14 Enlarged lymphatic vessels

Angiopoietin-1 25

Growth factor Tg/K14 Lymphatic vessel enlargement and sprouting

HGF 26 Growth factor Tg Increased number and enlargement of lymphatic vessels

1(Wigle et al. 2002; Harvey et al. 2005). 2(Huang et al. 2006; Gale et al. 2007). 3(Karkkainen et al. 2004). 4(Dumont et al. 1998). 5(Abtahian et al. 2003; Sebzda et al. 2006). 6(Taniguchi et al. 2007). 7(Backhed et al. 2007). 8a(Schacht et al. 2003). 8b(Fu et al. 2008) 9(Petrova et al. 2004; Seo et al. 2006). 10(Makinen et al. 2005). 11(Yuan et al. 2002). 12(Gale et al. 2002; Shimoda et al. 2007). 13(Hirashima et al. 2008). 14(Fritz-Six et al. 2008). 15(Danussi et al. 2008). 16(Huang et al. 2000). 17(Ichise et al. 2009). 18(Kulkarni et al. 2009). 19(Ayadi et al. 2001). 20(Francois et al. 2008). 21(Gupta et al. 2007). 22(Jeltsch et al. 1997). 23(Veikkola et al. 2001). 24(Kunstfeld et al. 2004). 25(Tammela et al. 2005). 26(Kajiya et al. 2005). Angptl4, angiopoietin-like protein 4; Aspp, apoptosis stimulating protein of p53; ECM, extracellular matrix; Foxc, forkhead box C; HGF, hepatocyte growth factor; K14, keratin 14; KO, knockout; LYVE-1, lymphatic vascular endothelial hyaluronan receptor-1; NRP, neuropilin; PDZ, PSD-95, DISCS-large, and ZO-1; SLP, Src homology 2-domain containing leukocyte protein; SOX, sex determining region Y-related high mobility group box; Spred, Sprouty-related Ena/VASP homology 1 domain-containing protein; Tg, transgenic; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor.

Page 29: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

28

2.1.4 Lymphatic vascular development in Xenopus Laevis

Amphibians have a common evolutionary history with mammals that is an estimated 100

million years longer than that between zebrafish and mammals. Being both tetrapods,

amphibians and mammals share extensive synteny at the level of the genomes and have many

similarities in organ development, anatomy, and physiology (Christensen et al. 2008; Raciti et

al. 2008). In the past, embryos and tadpoles of the African clawed frog (Xenopus laevis) have

served as a powerful animal model to study blood vascular development and angiogenesis

(Cleaver and Krieg 1998; Helbling et al. 2000; Levine et al. 2003; Kalin et al. 2007). More

recently, Xenopus embryos were shown to develop also a complex, well-defined lymphatic

vascular system (Ny et al. 2005). Similar to the development of the mammalian lymphatic

vascular system, in Xenopus, LECs transdifferentiate from venous cells BEC and

lymphangioblasts contribute to newly forming lymph vessels that mature to drain fluids from

the peripheral tissues back to the blood circulation. The lymphatic vascular system of the

Xenopus, in contrast to the mammalian one, presents a lymph heart.

2.1.4.1 Rostral lymph sac, lymph heart and caudal lymphatic vessel formation

Prox1 expression is first found in the head from stage 28 on. Later on, cells of the lateral plate

mesoderm co-express Prox1, VEGFR-3 (that is expressed in the whole developing vasculature)

and angioblast markers (e.g mesenchyme-associated serpentine receptor (Msr)). Once the

Prox1+ cells became restricted to the subectodermal regions, Msr expression is lost and the

Prox1+/VEGFR-3+/Msr- population is now defined as lymphangioblasts; these cells will

assemble and form the rostral lymph sac (Ny et al. 2005).

Similar to the mammalian system, at stage 32, a subpopulation of the venous endothelial cells in

the anterior trunk start expressing Prox1 and progressively lose Msr expression; they

Page 30: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

29

transdifferentiate into the lymphatic lineage and form the lymph heart. Furthermore, Prox1

expression is detected in the posterior trunk. Prox1+ lymphatic endothelial cells lining the

posterior cardinal veins detach from it and form the ventral caudal lymph vessel. Sometimes

even before the posterior cardinal veins have acquired a lumen, Prox1+ endothelial cell clusters,

the putative lymphangioblast, appear. At stage 33/34, the expression of LYVE-1 is weakly

detected in all vessels. At stage 35, VEGFR-3 expression begins to be restricted to the

lymphatic vessels while LYVE-1 expression is restricted to the lymphatic vessel only from

stage 39. From stages 37/38 onward, endothelial cells of the dorsal longitudinal anastomosing

vessel, a vein draining blood to the heart, were also found to transdifferentiate into Prox1+ cells.

As a result, (by stage 42) a lumenized ventral and dorsal caudal vein are formed. Thus, venous

transdifferentiation and lymphangioblast differentiation contribute to the formation of the caudal

lymph vessels. A schematic representation of the early stages of lymphatic vascular system

development (Prox1 expression) in Xenopus tadpoles is depicted in Figure 2.1.4

Figure 2.1.4 Expression of Prox1 and Msr in early tadpoles (from Ny et al. 2005). (a-d) in situ hybridization

(blue) for Prox1 (left panel, lymphatic vessels) and Msr (righ panel, blood vessels). Prox1 is expressed in the head

where the rostral lymph sac develops (RLS); in the anterior trunk adjacent to the pronephric sinus (PNS), where the

lymph hearth forms (LH); and in the posterior trunk adjacent to the posterior cardinal vein (PVC) where the ventral

Page 31: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

30

caudal lymph vessel (VCLV) and dorsal caudal lymph vessel (DCLV) form. Prox1 is also expressed in the eyes (E)

and liver diverticle (L). Later in development additional commitment sites emerge from the dorsal longitudinal

anastomosing vessel (DLAV). Msr is expressed in the dorsal aorta (DA), in the cardinal vein, in the intersomitic

veins (ISV), in the heart (H) and in the aortic arch artery (AAA). Scale bars: a-c, 1 mm; d, 500 µm.

2.1.4.2 Molecular mechanisms of the lymphatic vascular development in Xenopus

Antisense-morpholino knockdown studies of the lymphatic master gene Prox1 revealed that

lack of this transcription factor impairs lymphatic development (and not blood vasculature

development) resembling the Prox1-/- mouse phenotype (Ny et al. 2005). In addition, antisense-

morpholino knockdown of the lymphangiogenic factor VEGF-C causes lymphatic vessel defects

similar to the phenotype observed in VEGF-C deficient mice, including impaired LEC sprouting

and migration, and the formation of lymphedema (Ny et al. 2005). Single morpholino antisense

knockdown of VEGF-D did not affect lymphatic commitment, but transiently impaired

lymphatic endothelial cell (LEC) migration. Notably, combined knockdown of VEGF-D with

VEGF-C resulted in more severe migration defects and lymphedema formation than the

corresponding single knockdowns. The stronger phenotype might be possibly due to the fact

that, in the single knockdowns, each factor might have compensated for the loss of the other.

Knockdown of VEGFR-3 or treatment with a VEGFR-3 inhibitor similarly impaired lymph

vessel formation and function and caused pronounced edema. VEGFR-3 silencing by

morpholino knockdown, inhibitor treatment, or VEGF-C/D double knockdown also resulted in

dilation and dysfunction of the lymph heart. These findings document a critical role of VEGFR-

3 in embryonic lymphatic development and function and reveal a previously unrecognized

modifier (VEGF-D null mice have no lymphatic phenotype (Ny et al. 2008)) role of VEGF-D in

the regulation of embryonic lymphangiogenesis in frog embryos (Ny et al. 2008). Recently,

using a genetic screen in zebrafish, ccbe1 (collagen and calcium-binding EGF domain-1) was

identified as indispensable for embryonic lymphangiogenesis. Ccbe1 acts at the same stage of

Page 32: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

31

development as VEGF-C and is required for the budding of the lymphangioblast and angiogenic

sprouting from venous endothelium (Hogan et al. 2009). Furthermore, a novel two-step

screening strategy involving a simple phenotypic read-out (edema formation or larval lethality)

followed by semi-automated in situ hybridization using a chemical library of 1,280 bioactive

compounds identified the adenosine A1 receptor antagonist, 7-chloro-4-hydroxy-2-phenyl-1,8-

naphthyridine, as an inhibitor of blood vascular and lymphatic development in Xenopus frogs

(Kalin et al. 2009).

2.1.5 Molecular markers of lymphatic endothelium

For many years, the lack of specific lymphatic markers has hampered research in the lymphatic

vascular field. However, the recent establishment of pure blood and lymphatic endothelial cell

populations isolated from human dermal tissues has helped improving our understanding on the

development and function of the lymphatic system (Kriehuber et al. 2001; Makinen and Alitalo

2002; Podgrabinska et al. 2002; Hirakawa et al. 2003). LECs and BECs maintain their lineage-

specific differentiation in culture even after several passages and gene expression profiling

experiments have revealed that almost 98% of the investigated genes were expressed at

comparable levels in both cell types (Petrova et al. 2002; Hirakawa et al. 2003). Interestingly,

this approach identified previously unknown lymphatic and blood vascular lineage-specific

markers (see Table 2.1.2)

Page 33: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

32

Table 2.1.2 Lymphatic and blood vascular lineage-specific markers

Markers Function LV BV References

Prox1 Transcription factor ++ (Wigle et al. 1999)

Podoplanin Transmembrane glycoprotein

++ (Wetterwald et al. 1996; Breiteneder-Geleff et al. 1999)

LYVE-1 Hyaluronan receptor ++ (Banerji et al. 1999)

VEGFR-3 Growth factor receptor + /(+)1 (Kaipainen et al. 1995)

Neuropilin-2 Growth factor receptor + /(+)2 (Yuan et al. 2002)

Macrophage mannose receptor-1

Adhesion molecule + (Irjala et al. 2001; Irjala et al. 2003)

CCL21 CC-chemokyne + (Gunn et al. 1998)

CCL20 CC-chemokyne (++)3 (++)3 (Kriehuber et al. 2001; Hirakawa et al. 2003)

Desmoplakin Anchoring protein of adherent junctions

+ (Ebata et al. 2001)

Plakoglobin Connect cadherins to cytoskeleton in cell-cell junction

+ (Petrova et al. 2002; Hirakawa et al. 2003)

Integrin α9 Adhesion molecule, VEGFR3 co-receptor

+ (Huang et al. 2000; Petrova et al. 2002)

CD44 Hyaluronan receptor + (Kriehuber et al. 2001)

VEGF-C Growth factor + (Kriehuber et al. 2001; Hirakawa et al. 2003)

VEGFR-1 Growth factor receptor + (Hirakawa et al. 2003)

Neuropilin-1 Growth factor receptor + (Hong et al. 2002; Petrova et al. 2002)

Endoglin TGF-b receptor ++ (Hirakawa et al. 2003)

CD34 Adhesion molecule /(+)4 ++ (Young et al. 1995)

IL-8 CXC-chemokine + (Petrova et al. 2002)

N-cadherin Adhesion molecule + (Petrova et al. 2002; Hirakawa et al. 2003)

ICAM Adhesion molecule + (Erhard et al. 1996)

Integrin α5 Adhesion molecule + (Petrova et al. 2002; Hirakawa et al. 2003)

Collagen IV ECM protein /(+)5 ++ (Hirakawa et al. 2003)

Versican Proteoglycan + (Petrova et al. 2002; Hirakawa et al. 2003)

Laminin Basement membrane molecule

/(+)5 ++ (Barsky et al. 1983; Petrova et al. 2002)

Collagen XVIII Basement membrane molecule

/(+)5 ++ (Petrova et al. 2002; Hirakawa et al. 2003)

Page 34: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

33

2.1.6 Molecular mediators of adult lymphangiogenesis

The lymphatic vasculature remains quiescent in a physiological adult state. However, during

tissue regeneration, wound healing, tumor growth and inflammation, new lymphatic vessels

develop from preexisting ones. In addition, circulating endothelial progenitor cells (EPCs) and

macrophages have been reported to transdifferentiate into LECs during chronic renal transplant

rejection and corneal lymphangiogenesis (Maruyama et al. 2005; Religa et al. 2005;

Schledzewski et al. 2006). In contrast, bone marrow-derived progenitor cells fail to incorporate

into the endothelium of newly formed lymphatic vessels in mouse tumor xenografts (He et al.

2004) and growing evidence suggests that these cells might only serve as supporting cells that

promote vessel growth (Grunewald et al. 2006; Purhonen et al. 2008).

VEGFR-3 is a potent mediator of adult lymphangiogenesis and VEGF-C and -D are presently

the only known ligands for this receptor (Figure 2.1.5). VEGF-C promotes proliferation,

migration and survival of LECs in vitro (Makinen et al. 2001). VEGF-D also stimulates

lymphangiogenesis in tissue and tumors (Stacker et al. 2001; Veikkola et al. 2001), and

transgenic mice overexpressing VEGF-C or -D in the skin, show hyperplasia of dermal

lymphatic vessels (Jeltsch et al. 1997; Veikkola et al. 2001). In addition, after proteolytic

cleavage, the fully processed, mature forms of VEGF-C and -D can bind to the major known

regulator of angiogenesis, VEGFR-2 (Joukov et al. 1997; Achen et al. 1998). Recently, it has

BV, blood vessels; CCL, CC chemokine ligand; LV, lymphatic vessel; LYVE-1, lymphatic vascular endothelial hyaluronan receptor-1; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor. 1VEGFR-3 expression was also found on same blood capillaries during tumor neovascularisation and in wound granulation tissue (Valtola et al., 1999; Paavonen et al., 2000). 2Neuropilin-2 is also expressed in veins (Yuan et al., 2002). 3After activation, both blood vascular and lymphatic endothelial cells strongly express CCL20 (Kriehuber et al., 2001). 4CD34 expression has also been found on lymphatic endothelial cells (Sauter et al., 1998; Kriehuber et al., 2001). 5Peripheral lymphatic vessels sometimes have incomplete basement membrane, large collecting vessels a complete one.

Page 35: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

34

been suggested that VEGFR-2 and -3 might cooperate in terms of LEC migration and

proliferation but not vessel formation (Goldman et al. 2007). However, a mutant VEGF-C,

unable of stimulating VEGFR-2, was shown to be sufficient to activate lymphangiogenesis

(Joukov et al. 1998; Veikkola et al. 2001). Thus, the distinct contribution of VEGFR-3 and 2

towards lymphangiogenesis remain, at present, unclear.

Neuropilins, non-kinase type I transmembrane proteins, are involved in axonal guidance.

Neuropilin-2 expression is also found in veins and lymphatic capillaries. It binds to VEGF-C

(Figure 2.1.5) and this has suggested the possibility that VEGFR-3 signalling could be enhanced

by NRP2 in a similar way as for NRP1 and VEGFR-2 (Soker et al. 2002). Furthermore, the

angiopoietin/Tie signalling pathway is essential for blood vasculature development and recently,

it has been shown that Ang1 also induces lymphangiogenesis in the mouse cornea (Morisada et

al. 2005) and after adenoviral gene transfer in other mouse tissues (Tammela et al. 2005). Ang1

is thought to act either directly on the lymphatic endothelium since its receptor Tie2 is

expressed in LECs in vitro and in vivo (Kriehuber et al. 2001; Petrova et al. 2002) or indirectly

via the VEGF-C-VEGFR-3 pathway, since Ang1 effect could be blocked with soluble VEGFR-

3 (Tammela et al. 2005b) (Figure 2.1.5). Ang2 is considered to be an antagonist of Tie2, but the

study of Ang2 null mice suggest that Ang2 might act as an agonist of Tie2 in

lymphangiogenesis while being an antagonist in angiogenesis (Gale et al. 2002).

Fibroblast growth factor (FGF)-2 (Chang et al. 2004) and hepatocyte growth factor (HGF) have

been identified as lymphangiogenic factors (Kajiya et al. 2005) promoting in vitro and in vivo

lymphangiogenesis via their receptors FGFR-3, (Shin et al. 2006) and HGFR (Kajiya et al.

2005) (Figure 2.1.5). In contrast to the FGF2 effect, HGF activity was not blocked by inhibiting

VEGFR-3 signalling (Kajiya et al. 2005), so HGF might act directly on the lymphatic

endothelium, while FGF2 could have an indirect effect (Kubo et al. 2002).

Additional lymphangiogenic factors have been proposed, including platelet derived growth

factor (PDGF)-BB (Cao et al. 2004), insulin growth factor (IGF)1-2 (Bjorndahl et al. 2005),

Page 36: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

35

growth hormone (GH) (Banziger-Tobler et al. 2008), adrenomedullin (AM) (Jin et al. 2008) and

sphingosine-1-phospate (S1P) (Yoon et al. 2008) (Figure 2.1.5). Conversely, transforming

growth factor (TGF)-β signalling negatively regulates lymphangiogenesis in inflamed tissues as

well as in certain tumor tissues (Oka et al. 2008).

Figure 2.1.5 Receptors expressed by lymphatic endothelium and lymphangiogenic growth factors (from

(Jurisic and Detmar 2009)). Activation of VEGF receptors-2 and -3 (VEGFR-2, VEGFR-3) and neuropilin-2

(Nrp2) by several vascular endothelial growth factors (VEGF-A, VEGF-C, VEGF-D) promotes lymphangiogenesis.

Additional lymphatic growth factors include angiopoietin-1 (ANG-1), hepatocyte growth factor (HGF), fibroblast

growth factor-2 (FGF-2), insulin-like growth factors (IGF-1, IGF-2), platelet-derived growth factor-BB (PDGF-

BB) and adrenomedullin (AM)

2.1.7 Involvement of lymphatic vessels in disease

2.1.7.1 Lymphedema

Lymphedema is the accumulation of tissue fluid as a direct consequence of a dysfunctional

lymphatic system, where abnormal or damaged lymphatic vessels are unable to drain fluid from

Page 37: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

36

the interstitium. Lymph accumulation is characterized by severe swelling of the affected limb,

fibrosis, degeneration of the connective tissue and a high risk of infections (Rockson 2001).

Lymphedemas have been divided into two main groups: primary and secondary.

Primary lymphedema has no identifiable cause and it can appear at birth (congenital), at puberty

(praecox) or in adulthood (tarda). Congenital lymphedema has been associated with a missense

mutation that encodes for a non active VEGFR-3 protein, in Milroy’s disease (Brice et al. 2005).

Lymphedema-distichiasis, that has been associated with mutations in the Foxc2 gene (Fang et

al. 2000; Finegold et al. 2001) is characterized by a double row of eyelashes at birth,

lymphedema at puberty, abnormal lymphatic patterning, agenesis of lymphatic valves, and

abnormal recruitment of SMCs (Petrova et al. 2004). Moreover, mutations of the SOX18 gene

have been described in hypotrichosis-lymphedema-telangiectasia syndrome (Irrthum et al.

2003). Finally, nuchal edema, lymphatic dysplasia and jugular lymph sac distension are

associated with a variety of human syndromes: Edwards syndrome (trisomy of chromosome 18,

(Bekker et al. 2006), Down’s syndrome (trisomy of chromosome 21), Turner’s syndrome (all or

part of one of the X chromosome is absent), Patau’s syndrome (trisomy of chromosome 13),

lethal multiple pterygium syndrome (reviewed in (Chitayat et al. 1989; Christensen et al. 2008)

and Noonan’s syndrome (mutation in the PTPN11, SOS1, KRAS and RAF1 genes or

duplication of chromosome region encompassing gene PTPN11) (Ferreira et al. 2008;

Malaquias et al. 2008). Nuchal edema has been also reported in Fryns syndrome, for which the

disease-causing mutations have not been yet identified (Slavotinek et al. 2005). In most of the

families with lymphedema, however, none of the known mutations occurs, thus several more

genes involved in lymphedema remain unidentified.

Secondary lymphedema develops when the lymphatic vasculature is damaged by infections or

surgery. Filariasis, a parasitic tropical disease, is the main cause of lymphedema (120 million

people affected) (Wynd et al. 2007). The cause of the infection are mosquito-borne parasites

Wuchereria bancrofti and Brugia malayi, which live and reproduce in the lymphatic system;

Page 38: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

37

thus the lymphatic vessels are damaged, no lymph fluid is transported and edema appears in the

legs and genitals (Melrose 2002). Current therapies try to target the worms but no drug

(ivermectin or doxycyclin) so far has been able to cure the disease. While the filariasis-

lymphedema is mostly present in tropical and subtropical countries, edema of the arm or of the

axilla after lymph node dissection is the main cause of lymphedema in the industrialized

countries. Lymphedema occurs in 6-30% of mastectomy patients and this rate increases if they

are exposed to radiotherapy. In the future, a combination of VEGF-C therapy and lymph node

transplantation would represent a novel treatment option for individuals with a history of cancer

(Tammela et al. 2007). Importantly, based on its potent lymphangiogenic effect, VEGF-C has

been tested for gene and protein therapy in animals. Adenoviral mediated-VEGF-C and VEGF-

C156S (a mutant protein that only activates VEGFR-3, (Saaristo et al. 2002a; Saaristo et al.

2002b)) gene therapy has been shown to promote lymphatic vessel generation in the skin

(Karkkainen et al. 2001). Furthermore, regeneration of lymphatic vessels was observed after

injection of VEGF-C in a surgical model of lymphedema in the rabbit (Szuba et al. 2002).

2.1.7.2 Lymphatic vessels and inflammation

Lymphatic vessels participate in inflammation by transporting leukocytes from the site of

inflammation to secondary lymphoid organs. Upon exposure to an inflammatory stimulus,

dendritic cells (DCs) capture antigens in peripheral tissues and migrate to lymph nodes. After

their maturation, DCs express higher levels of the lymphoid chemokine receptor CCR7

(Sallusto et al. 1998) which promotes their migration into lymph nodes, attracted by the

chemokine CCL21 (or SLC, secondary lymphoid tissue chemokine produced by LECs)

(Sallusto et al. 1998; Saeki et al. 1999; Ohl et al. 2004). Once in the lymph node, antigenic

peptide-loaded DCs mediate the presentation of major histocompatibility complex (MHC)-

peptide complexes to T cells that become activated (Bajenoff et al. 2003). DCs and T cells leave

Page 39: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

38

peripheral tissues, enter the lymphatic vessels and migrate towards lymph nodes also via

interaction of CCR7 (Bromley et al. 2005; Debes et al. 2005) with adhesion molecules

expressed by lymphatic endothelium such as CLEVER-1 (common lymphatic endothelial and

vascular receptor-1) (Salmi et al. 2004) and mannose receptor 1 (Irjala et al. 2001). Interaction

of sphingosine-1-phosphate (S1P) with its receptor S1P1 has been implicated in the exit of

lymphocytes from the lymphnodes (Matloubian et al. 2004; Mandala et al. 2002). VEGF-C has

been shown to be upregulated in response to proinflammatory cytokines, presumably through

NF-κB-mediated promoter activation (Ristimaki et al. 1998). Together with VEGF-A, which is

highly expressed by follicular B cells, VEGF-C is a possible mediator of the increased

lymphangiogenesis and DC migration (Angeli et al. 2006). Inflammation-induced

lymphangiogenesis can also be mediated by macrophages (Kerjaschki 2005). They are recruited

into the inflammation site and can transform from naive monocytes into VEGF-C/D-producing

cells (Schoppmann et al. 2002). Human kidney transplant rejection is frequently accompanied

by lymphangiogenesis, and CCL21 produced by the LECs might actively promote the

inflammatory process (Kerjaschki et al. 2004). Furthermore, VEGF-A mediated lymphatic

hyperplasia is observed in UVB-induced skin inflammation in human psoriatic skin lesions

(Kunstfeld et al. 2004; Kajiya et al. 2006). Mice overexpressing VEGF-A in the skin display a

prolonged inflammatory response after induction of delayed-type-hypersensitivity (DTH),

which is associated with LEC proliferation and lymphatic hyperplasia and can be inhibited by

blockade of VEGFR-1 and -2 (Kunstfeld et al. 2004). In chronic airway inflammation induced

by Mycoplasma pulmonis, massive lymphangiogenesis is induced by VEGF-C/-D-producing

inflammatory cells, while blocking VEGFR-3 signalling resulted in bronchial lymphedema

(Baluk et al. 2005). Finally, the role of VEGF-C/D–VEGFR-3 signalling in inflammatory

lymphangiogenesis is also suggested by overexpression of VEGF-C in the joint synovium of

rheumatoid arthritis patients (Paavonen et al. 2002). Together, these studies underline the close

relationship between inflammation, the immune response and lymphangiogenesis.

Page 40: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

39

2.1.7.3 Tumor lymphangiogenesis and metastasis

A major prognostic indicator for the progression of human cancers is the detection of metastasis

in regional lymph nodes. Primary cancer cells invade local tissue, enter tumor-associated blood

vessels as a means to target distant organs and enter tumor-associated lymphatic vessels to

target draining (sentinel) distal lymph nodes and distal organs. The lymphatic pathway is the

most common pathway of initial cancer cell dissemination (reviewed by (Stacker et al. 2002a;

Stacker et al. 2002b). Despite this evidence, the molecular mechanisms of lymphatic metastasis

are not completely understood.

Tumor cells can invade pre-existing lymphatic vessels in the surrounding tissues or intratumoral

lymphatic networks (Karpanen et al. 2001; Mandriota et al. 2001; Skobe et al. 2001; Stacker et

al. 2001; Cao et al. 2004; Achen et al. 2005; Cao 2005b; Cao 2005a). In some cases of human

cancers, the presence of intra-tumoral lymphatic vessels was reported to correlate positively

with lymph node metastasis and poor prognosis (Beasley et al. 2002; Dadras et al. 2005; Kyzas

et al. 2005).

Importantly, tumor cells and macrophages secrete growth factors, such as VEGF-C and -D that

can facilitate the transmigration of malignant cells through the lymphatic endothelium.

(Schoppmann et al. 2002), VEGF-C stimulates the growth of new tumor-associated lymphatic

vessels, and in this process nearby LECs send filopodia toward VEGF-C producing tumor cells,

and then form tumor-directed vessel sprouts, where the vessel lumen opens up and allows

facilitated access of tumor cells (He et al. 2005). Moreover, intraluminal VEGF-C also promotes

the dilation of collecting lymphatic vessels, endothelial proliferation in the vessel wall and

further enhances the delivery of tumor cells to sentinel lymph nodes, probably by increasing the

lymph flow rate (He et al. 2005; Hoshida et al. 2006).

Page 41: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

40

Direct experimental evidences that increased levels of VEGF-C or VEGF-D promote active

tumor lymphangiogenesis and lymphatic tumor spread to regional lymph nodes have been

provided by studies in animal tumor models (Karpanen et al. 2001; Mandriota et al. 2001;

Skobe et al. 2001; Stacker et al. 2001). These effects are suppressed by specific inhibition of the

VEGFR-3 signalling pathway by either VEGFR-3 blocking antibody, VEGF-C/D trap

(VEGFR-3 extracellular domain fused with immunoglobulin Fc portion) or VEGF-C targeting

siRNA. This inhibition prevents cancer metastasis to lymph nodes and distant organs (Karpanen

et al. 2001; He et al. 2002; Lin et al. 2005; Roberts et al. 2006), thus underlining the association

of VEGF-C or VEGF-D expression and metastasis. Combination of siRNA therapy targeting

both VEGF-C and VEGF-A inhibits lymph node and lung metastasis in a mouse

immunocompetent mammary cancer model (Shibata et al. 2008). Moreover, because VEGF-C

can also activate VEGFR-2 and because VEGF-A promotes lymphangiogenesis, also blocking

VEGFR-2 results in moderate suppression of tumor lymphangiogenesis, as well as

angiogenesis, and a combination of anti-VEGFR-2 and -3 antibodies more potently decreases

lymph node and lung metastasis (Roberts et al. 2006). Recently, it has been shown that

endothelial nitric oxide synthase (eNOS) mediates VEGF-C-induced lymphangiogenesis in

mice; this finding explains the correlation between eNOS and lymphatic metastasis seen in a

number of human tumors and could open the door for potential therapies targeting NO

signalling (Lahdenranta et al. 2009). Moreover, neuropilin-2 expression has been detected on

tumor activated, but not on quiescent, lymphatic vessels (Aslakson and Miller 1992; Caunt et al.

2008). A blocking antibody targeting neuropilin-2 inhibited tumor lymphangiogenesis and

reduced lymphatic metastasis, leaving quiescent mouse lymphatics unaffected (Caunt et al.

2008).

As said, migration to regional lymph nodes represents the first step of tumor metastasis in many

cancers and is an important prognostic indicator for progression of the disease. Interestingly, in

Page 42: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

41

animal tumor models it has been shown that primary tumors can prepare a favourable

environment for their future metastatic site even prior to their arrival, partly by producing

lymphangiogenic factors such as VEGF-A (Hirakawa et al. 2005) and VEGF-C (Hirakawa et al.

2007) that promote lymphatic vessel growth in the sentinel lymph nodes. Tumor-infiltrating

macrophages could also contribute to these processes by secreting lymphangiogenic factors

(Schoppmann et al. 2002). Clinicopathological studies of human cancers have shown a direct

correlation between expression of VEGF-C or VEGF-D by tumor cells and lymphatic invasion,

lymph node and distant metastasis, and poor patient survival (Stacker et al. 2002b; Pepper et al.

2003; Achen et al. 2005; Tobler and Detmar 2006). The lymphatic endothelium may also

actively participate in metastasis formation by secreting chemokines such as CCL21, whose

receptor is expressed on some malignant cells (Zlotnik 2004; Shields et al. 2007). Collectively,

these findings suggest that inhibition of lymphangiogenesis at the site of the primary tumor as

well as in the draining lymph node might be of interest for preventing tumor metastasis.

At last, it is important to shortly mention that a role for the lymphatic vascular system in obesity

has been, recently suggested. In fact, fat is accumulated in the edematous tissues of

lymphedema patients and mice lacking one allele of the Prox1 gene develop adult-onset obesity

due to abnormal lymph leakage (Harvey et al. 2005). In addition, a role for lymphatic capillaries

and VEGF-C signalling has also been proposed in hypertension (Machnik et al. 2009). Further

studies will definitely characterize these two processes in the near future.

Page 43: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

42

2.2 Mouse embryonic stem cell-derived embryoid bodies

Murine embryonic stem (ES) cells are pluripotent cells capable of differentiating into all

somatic and germ cell types. They are derived from the inner cell mass (ICM) of mouse

blastocysts. When cultured in the presence of anti-differentiation agents such as leukemia

inhibitory factor (LIF) and embryonic fibroblast feeder-cell layers (PMEFs), these cells

maintain their pluripotency and proliferate (Evans and Kaufman 1981) (Figure 2.2.1)

Figure 2.2.1 Mouse embryonic stem cell isolation and culture

(modified from (Wobus and Boheler 2005)). Mouse embryonic

stem (ES) cells are obtained by isolation of the inner cell mass

(ICM) from mouse blastocysts. Once isolated the ES cells can be

cultured in vitro for several passages.

Removal of LIF and PMEFs causes the ES cells to spontaneously differentiate, thus

recapitulating early embryogenesis. When ES cells are cultured in suspension without anti-

differentiation agents, they form three-dimensional aggregates called embryoid bodies (EB).

Within 2-4 days in suspension culture, endoderm forms from the inner cell mass, thereby giving

rise to simple EBs (Leahy et al. 1999). On approximately day 4, differentiation of columnar

epithelium with a basal lamina and formation of a central cavity occurs. At this stage, the EBs

are called cystic embryoid bodies (Evans and Kaufman 1981). Upon continued in vitro culture,

EBs give rise to all three germ layers (ectoderm, mesoderm, endoderm) and terminally

Page 44: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

43

differentiate into a wide variety of cell types, such as cardiomyocytes, hematopoietic cells,

neurons, pancreatic islet cells blood vascular and lymphatic endothelial cells (Risau et al. 1988;

Bain et al. 1995; Dang et al. 2002; Liersch et al. 2006). The in vitro culture of ES cells as EBs

affords opportunities to mechanistically study early differentiation events of 3D assemblies of

pluripotent cells. One advantage of in vitro differentiation studies is that ES cells can be studied

for gene mutations or knockouts that prove to be lethal during normal embryonic development

in vivo.

Global DNA microarray analysis indicates that EBs temporally express genes in a manner that

recapitulates the sequence of normal development from primitive ectoderm formation, to

gastrulation (Figure 2.2.2), and eventual early cell specification prior to organogenesis (Dvash

et al. 2004). EBs express most, if not all, factors necessary to induce and regulate

embryogenesis (Czyz and Wobus 2001; O'Shea 2004). Expression of phenotypic markers of

endoderm (such as Foxa2, Sox17, GATA 4/6, -fetoprotein, and albumin), mesoderm (such as

brachyury-T, Msp1/2, Isl-1, -actin, -globin, and Runx2), and ectoderm (such as Sox1, nestin,

Pax6, GFAP, Olig2, neurofilament, and -III tubulin) demonstrate the ability of EBs to generate

cells from all three germ layers (Itskovitz-Eldor et al. 2000). Furthermore, comparison of the

morphology of an egg cylinder stage mouse embryo (ca. embryonic day 6) with an attached day

7 EB suggests that early stage EBs do mimic early mouse embryogenesis in an organized

manner (Figure 2.2.2). However, time-wise it may very well be that embryonic–like in vitro

development is shifted or spread over time and space; in fact, continuous development of new

cells might disturb the differentiation processes.

Page 45: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

44

Figure 2.2.2 Schematic representation of cells and

structures of an embryoid body on day 7 and an

early egg stage cylinder mouse embryo (ED 6). Both

present a parietal (yellow) and visceral (blue)

endoderm, primitive ectoderm (green) and mesoderm

(red). The EB does not present a trophoectoderm (light

pink). The EB central cyst represents the proamniotic

cavity while the blastocoel in the mouse embryos is

substituted with culture medium (pink stripes)

2.2.1 EB culture methods

Common EB culture practices include hanging drop and static suspension culture. The hanging

drop method of EB formation produces homogeneous cell aggregates by dispensing a defined

number of ES cells in physically separated droplets of media suspended from the lid of a Petri

dish (Hopfl et al. 2004). In contrast, static suspension culture is performed by simply adding a

suspension of ES cells to a bacteriological grade Petri dish, thereby allowing the cells to

spontaneously aggregate via cell–cell adhesions (Zhang et al. 2001). Static suspension cultures

produce a large number of EBs rather simply, but the size and shape of the resulting EBs are

highly variable due to the tendency of EBs to agglomerate after initial formation in static

suspension, often producing large, irregularly shaped masses of cells. Entrapment of a single

cell suspension or small clusters of ES cells in hydrogels, such as hyaluronic acid (Gerecht et al.

2007) represents a compromise between hanging drop and static suspension approaches to attain

physically separated EBs in a bulk semi-solid suspension medium; however, the efficiency of

EB formation from individual ES cells can be rather low. Alternative techniques for EB

Page 46: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

45

formation and culture have also been developed recently, using multiwell and microfabrication

technologies. These induce aggregation more rapidly than hanging drops but still require

individual processing and manipulation of the resulting EBs (Ng et al. 2005).

2.2.2 Vasculogenesis, blood and lymphatic vascular development and

(anti-) lymph-angiogenesis research

Vasculogenesis is the formation of blood vessels by in situ differentiation of angioblast

precursors. Taking place right after gastrulation, it is one of the earliest and essential

differentiation processes during in vivo embryogenesis. EBs are avascular for the first 4 days of

development. This results in a gradient of oxygen and a deficit of nutrients toward the center of

the EB. As a consequence, hypoxia inducible factor-1α and concomitant VEGF-A become

upregulated, initiating vasculogenesis. At day 5 of culture, clusters of Flk1+, CD31+ (pecam),

MECA-32+, VE-cadherin+ and CD34+ endothelial cells are visible (Vittet et al. 1996). The

process of vasculogenesis is followed by angiogenesis, meaning proliferation and sprouting of

endothelial cells and vessel formation. Angiogenesis starts at day 8 of culture and 1-2 days are

needed to visualize the first blood vessel-like structures in the EBs (Vittet et al. 1996).

While the development of the blood vascular system has been long investigated using mouse

embryoid bodies, the differentiation of ES into lymphatic endothelial cells has been reported

only recently (Figure 2.2.3, (Liersch et al. 2006)). In the EBs, the lymphatic vessel-like

structures develop after the formation of the blood vessel-like structure networks, thus

mimicking the in vivo situation (Figure 2.2.4).

Page 47: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

46

Figure 2.2.3 Development of lymphatic vessel-like structures in ES-derived EBs (modified from (Liersch et

al. 2006)). Double immunofluorescence stains of 21 days old EBs for CD31 (red; A, D), LYVE-1 (green, B;

arrows) and Prox1 (green, E; arrows) reveal CD31+ blood vessels and CD31+/LYVE-1+/Prox1+ lymphatic vessels

(C, F: merged images; arrows). Blood vessels expressed the blood vascular-specific marker MECA-32 (red, G)

whereas Prox1+ lymphatic vessels (H; green; arrow) were MECA-32- (I: merged image; arrow). Scale bars: 50 µm

Figure 2.2.4 Lymphatic vascular system development in mouse EBs largely mimics the in vivo situation. In

mouse embryos at embryonic day (ED) 5 the first progenitor endothelial cells develop. Later, the first blood vessels

and the heart form. At ED 9, as soon as the blood vessels mature, lymphatic competence and lymphatic

commitment are acquired by the endothelial cells of the anterior cardinal veins. Those lymphatic endothelial cells

will form the lymphatic vessels. In the embryoid bodies, both blood and lymphatic vessel-like structure formation

is delayed in time but the order of the in vivo developmental events is maintained (Liersch et al. 2006).

Page 48: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

47

The evidence that CD31+/LYVE-1+/Prox1+ lymphatic endothelial cells can differentiate and

form lymphatic vessel-like structures in the EBs, and the fact that they respond to

lymphangiogenic factors (e.g. VEGF-C, (Kreuger et al. 2006; Liersch et al. 2006)) has opened a

plethora of possibilities to better understand the mechanisms that control lymphatic vascular

development and, in particular, lymphatic endothelial differentiation. To this end, we have

developed an EB-lymphatic vascular differentiation assay that is depicted in Figure 2.2.5

Figure 2.2.5 Embryoid body (EB) lymphatic

differentiation and (anti-) lymphangiogenic assay. Mouse

embryonic stem cells colonies (red) are cultured in presence

of leukemia inhibitory factor (LIF) and a feeder layer of

mouse embryonic fibroblasts (grey). Then LIF and fibroblasts

are removed and single cells (yellow) are plated on a Petri

dish for suspension culture. After 3 days, EBs are formed

(green). EBs of same size are singularly picked and

transferred to 12multi well plates. After 14 days, treatment is

applied to the EBs (pink, yellow and blue wells). After 4

days, EBs are fixed and immunofluorescence staining (IF) is

performed for blood and lymphatic endothelial markers and

results are quantified.

Given the capacity of the EBs to form blood vessel-like structure, this in vitro model has been

used to screen for anti-angiogenic agents. For example, suramin, tamoxifen and

tetrahydrocortisol have been shown to inhibit angiogenesis in the EB system (Wartenberg et al.

1998; Wartenberg et al. 2001). As the lymphatic endothelial cells can differentiate and form

lymphatic vessel-like structures in the EBs, this in vitro model could serve to screen for novel

(anti)-lymphangiogenic agents as well. To this end, we have also developed an (anti)-

lymphangiogenic screening (Figure 2.2.5)

Page 49: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

48

3 AIM AND OUTLINE OF THIS DISSERTATION

The aim of the present work was to identify the molecular mechanisms that regulate the earliest

phases of lymphatic vascular system development during embryogenesis, as well as novel

lymphangiogenic pathways. Revealing the molecular processes that control lymphatic vessel

formation and remodeling might open up new possibilities for the treatment of lymphatic vessel

associated pathologies, such as lymphedema and cancer metastasis.

We have used mouse embryonic stem cell-derived embryoid bodies that largely mimic mouse

embryogenesis, to identify novel molecular players regulating lymphatic competence and

commitment, which represent the earliest events in lymphatic vascular system development.

This approach identified all-trans-retinoic acid (RA) and, more potently, in combination with

cAMP, as inducer of LYVE-1 (competence) and Prox1 (commitment) expression. In situ and in

vivo studies on mouse and Xenopus embryos confirmed that RA upregulates, while a RARα

inhibitor (Ro 41-5253) or a PkA inhibitor (H89) downregulates, the expression of LYVE-1 and

Prox1 in the endothelial cells of the cardinal veins and developing lymphatics (Chapter 4.1).

The second part of this thesis has focused on the identification of novel anti-lymphangiogenic

factors. Mouse embryoid bodies and human lymphatic endothelial cell in vitro assays were used

to investigate anti-lymphangiogenic properties of active compounds. The screening identified

GW2974, a small molecule inhibitor of the EGFR/ErbB2 tyrosine kinase, as an inhibitor of

lymphatic vessel formation. Moreover, human lymphatic endothelial cell-based in vitro and

mouse in vivo studies have suggested the EGFR/ErbB2 signalling pathway as a novel regulator

of lymphangiogenesis (Chapter 4.2).

Page 50: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

49

4 Identification of molecular mechanisms controlling

lymphatic vessel formation

4.1 A role for all-trans-retinoic acid in the early steps of

lymphatic vasculature development

4.1.1 Abstract

Despite recent progress in identifying lymphatic lineage-specific markers and growth factors,

the molecular mechanisms that regulate the earliest steps of lymphatic vascular system

development are unknown. To identify potential regulators of lymphatic competence and

commitment, we used an in vitro vascular differentiation assay with mouse embryonic stem

cell-derived embryoid bodies (EBs). We found that several growth factors, including VEGF-C,

growth hormone, insulin-like growth factor-1 and interleukin-7 moderately induced the

expression of the lymphatic competence marker LYVE-1 in the EBs. Importantly, incubation

with retinoic acid (RA) and, more potently, with RA in combination with cAMP, induced

LYVE-1 expression in the vascular structures of the EBs. This RA effect was dependent on

retinoid acid receptor (RAR)- and protein kinase A signalling. RA and cAMP incubation also

promoted the development of CD31+/LYVE-1+/Prox1+ cell clusters that arose independently

from vessel structures. In situ studies revealed that RAR-is expressed by endothelial cells of

the cardinal vein from ED 9.5 to 11.5 in mice, in areas of LYVE-1 expression. Timed exposure

of mouse embryos and Xenopus laevis tadpoles to excess of RA upregulated LYVE-1 and

VEGFR-3 on embryonic veins and increased formation of Prox1-positive lymphatic progenitors

and lymph sacs. These findings indicate that RA signalling mediates the earliest steps of

lymphatic vasculature development.

Page 51: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

50

4.1.2 Introduction

The lymphatic vascular system has an important role in the maintenance of tissue fluid

homeostasis, intestinal lipid absorption, and immune surveillance in that it recruits and

transports immune cells from peripheral tissues to the regional lymph nodes (Alitalo et al. 2005;

Cueni and Detmar 2008). There have been many studies into the role of the lymphatic system in

promoting metastasis to lymph nodes and beyond (Hirakawa et al. 2005; Hirakawa et al. 2007;

Rinderknecht and Detmar 2008) and in modulating inflammatory diseases (Kerjaschki 2006;

Jurisic and Detmar 2009). Much progress has been made following the identification of specific

lymphatic markers that distinguish lymphatic endothelial cells from blood vascular endothelial

cells and novel molecular mediators of lymphatic vessel growth and differentiation, as well as

developmental studies in mouse models (Alitalo et al. 2005; Cueni and Detmar 2008).

There is considerable evidence that during mammalian embryonic development, the lymphatic

vascular system predominantly develops from pre-existing embryonic veins. In mice, expression

of the lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1) at embryonic day (ED) 9-

9.5 by the endothelial cells that line the anterior cardinal veins is considered to be the first

morphological indication that venous endothelial cells have acquired the competence to respond

to an unidentified lymphatic-inducing signal (Oliver 2004). The biological function of LYVE-1

is unknown; LYVE-1–deficient mice have no major lymphatic or other abnormalities (Huang et

al. 2006; Gale et al. 2007). At approximately ED 9.5, a restricted subpopulation of endothelial

cells on one side of the cardinal vein express the transcription factor Prox1, indicating that this

is the stage of lymphatic commitment (Wigle et al. 2002). These Prox1-positive cells then bud

off from the cardinal veins and migrate away to finally form the primitive lymph sacs. Prox1-

deficient mice completely lack a lymphatic vascular system (Wigle et al. 2002). Vascular

budding and migration appear to occur under the guidance of local gradients of vascular

endothelial growth factor-C (VEGF-C), which activates its receptor, VEGFR-3, on lymphatic

Page 52: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

51

precursor cells (Karkkainen et al. 2004). VEGF-C–deficient mice also lack lymphatic

vasculature and undergo prenatal death because of pronounced fluid accumulation in the tissues

(Karkkainen et al. 2004). It was recently, shown that the transcription factor Sox18 is expressed

in a subset of the cardinal vein cells that later become Prox1-positive lymphatic progenitor cells

and that Sox18 directly activates Prox1 transcription (Francois et al. 2008).

Studies in genetic mouse models indicate that following the formation of the lymph sacs, the

separation of the lymphatic and venous system is mediated by the tyrosine kinase Syk and the

adaptor protein SLP-76 (Abtahian et al. 2003; Sebzda et al. 2006), the sprouty-related

ena/VASP homology 1 domain-containing proteins (spred)-1 and 2 (Taniguchi et al. 2007), and

angiopoietin-like protein 4 (Backhed et al. 2007). Further lymphatic vessel maturation and

remodeling are controlled by a plethora of molecules (reviewed by (Cueni and Detmar 2008))

that includes the transcription factor Foxc2 (Petrova et al. 2004), angiopoietin-2 (Gale et al.

2002; Shimoda et al. 2007), the non-kinase receptor neuropilin-2 (Yuan et al. 2002), ephrin B2

(Makinen et al. 2005) and the transmembrane glycoprotein podoplanin (Schacht et al. 2003).

Despite advances in our understanding of lymphatic vasculature development, the molecular

mechanisms that control the earliest stages of lymphatic competence (expression of LYVE-1 by

endothelial cells of the cardinal vein and lymphatic precursor cells) have not been determined.

To identify pathways that mediate lymphatic competence, we used a previously established

embryoid body (EB)-based vascular differentiation assay (Liersch et al. 2006) as a screening

model. This system assesses the ability of mouse EB cells to differentiate into lymphatic vessel-

like structures that express the panvascular marker CD31, as well as Prox1 and LYVE-1

(Liersch et al. 2006); it was previously used to characterize the ability of VEGF-C to promote in

vitro lymphangiogenesis (Kreuger et al. 2006; Liersch et al. 2006). Using this model system, we

Page 53: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

52

investigated the potential effects of soluble factors that have been previously reported to be

potentially associated with activity on lymphatic endothelial cells in vitro or in vivo.

In our study, the growth factors VEGF-C, growth hormone, insulin-like growth factor (IGF)-1

and interleukin (IL)-7 were found to moderately induce the expression of LYVE-1 in EBs.

Incubation of EBs with retinoic acid (RA) and, more potently, a combination of RA and

cyclicAMP (cAMP) induced LYVE-1 expression in the vascular structures; this effect is

dependent on retinoid acid receptor (RAR)- and protein kinase A (PKA) signalling. In situ

studies revealed that RAR-is highly expressed by endothelial cells of the cardinal vein from

ED 9.5–11.5 in mice, in areas of LYVE-1 expression. Most importantly, timed exposure of

mouse embryos and of Xenopus laevis tadpoles to RA resulted in potent upregulation of LYVE-

1 and VEGFR-3 on embryonic veins and lymph sacs. Together, these findings indicate that RA

signalling could mediate the earliest steps of lymphatic vasculature development.

4.1.3 Materials and methods

4.1.3.1 Mouse embryonic stem cell culture, establishment and treatment of EBs

Murine C57BL/6x129SvEv-derived (passage 3–12) embryonic stem cells (mES cells; kindly

provided by N. Gale, Regeneron Pharmaceuticals, Tarrytown, NY, USA), were cultured on

mitotically inactivated primary mouse embryonic fibroblasts (PMEFs, passage 2-5, Institute of

Laboratory Animal Science,University of Zurich, Switzerland) in Dulbecco’s modified Eagle

medium (Gibco, Eggenstein, Germany), supplemented with 18% fetal bovine serum (Gibco),

100 nM sodium pyruvate (Sigma), minumum essential medium (MEM) vitamins, 2 mM L-

glutamine, streptomycin and penicillin (all from Gibco), 10 mM 2-mercaptoethanol and 2000

U/ml recombinant leukaemia inhibitory factor (LIF; Chemicon International, Temecula, CA).

PMEFs and LIF were removed and mES cells were transferred to suspension culture for EB

formation as described (Kurosawa 2007). After 3 or 4 days, EBs of the same size were

Page 54: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

53

transferred into 12-well dishes (BD Bioscience, San Diego, CA) and cultured for 14 days

without LIF. Then, EBs were incubated with or without the following factors for 2, 4, 6, 8, 10,

12 or 14 days: 20 ng/ml recombinant human vascular endothelial growth factor-A (VEGF-A

165; kindly provided by the National Cancer Institute, Bethesda, MD); 200 ng/ml recombinant

human VEGF-C (R&D Systems, Minneapolis, MN); 20 ng/ml human fibroblast growth factor-2

(FGF-2, kindly provided by the National Cancer Institute); 100 ng/ml recombinant human

insulin growth factor-1 (IGF-1, R&D Systems); 25 ng/ml recombinant human interleukin-3 (IL-

3; Chemicon International); 30 ng/ml human hepatocyte growth factor (HGF, R&D Systems);

20 ng/ml human platelet growth factor (PLGF, R&D Systems); 50 ng/ml human growth

hormone (R&D Systems); 20 ng/ml recombinant human interleukin-7 (IL-7, Chemicon

International); 100 µM S-nitroso-N-acetylpenicillamine (SNAP, Sigma-Aldrich, Buchs,

Switzerland); 10 mM human endothelin-3 (ET3, R&D Systems); 1, 2,5, 5 ,10, or 100 µM all-

trans-RA (RA; Sigma); 10 µM 4-[E-2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)-

1-propenyl]benzoic acid (TTNPB, Sigma); 1 or 10 µM 13-cis-retinoic acid (13-cis-RA, Sigma);

0.5 mM 3',5'-cyclic monophosphate (cAMP; Fluka, Buchs, Switzerland); 10 µM Ro 41-5253

(BioMol International, Plymouth Meeting, PA); 10 µM N-[2-(p-bromocinnamylamino)ethyl]-5-

isoquinoline sulfonamide, Di-HCl salt (H89, CalbioChem, San Diego, CA). EBs were fixed in –

20°C cold 100% methanol or in 4% paraformaldehyde (PFA) at 4°C for 10 minutes.

4.1.3.2 Endothelial cell culture, real-time RT-PCR, FACS and immunostains

Human umbilical vein endothelial cells (HUVECs), obtained from ScienceCell Research Labs

(San Diego, CA) were seeded into fibronectin-coated culture dishes (10 µg/ml; BD Biosciences,

Bedford, MA) and were cultured in endothelial cell basal medium (EBM; Cambrex Bio Science,

Walkersville, MD) supplemented with 20% fetal bovine serum (FBS; Invitrogen, Grand Island,

NY), 2 mM L-glutamine, antibiotic-antimycotic solution, 10 µg/ml hydrocortisone and N6,2'-O-

Page 55: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

54

dibutyryl-adenosine 3',5'-cyclic monophosphate (25 µg/ml; all from Fluka, Buchs, Switzerland).

Cells from passages 6 to10 were used. HUVECs were incubated for 24 hours with 10 µM RA

plus 0.5 mM cAMP or with dimethyl sulfoxide (DMSO) as negative control. Then real-time

RT-PCR, fluorescence-activated cell sorting (FACS) and immunostaining were performed. For

real-time RT-PCR analyses, total cellular RNA was isolated using the Trizol reagent

(Invitrogen) and was extracted with chloroform, precipitated with isopropanol, washed with

70% ethanol, and dissolved in DNase-free/RNase-free distilled water. The concentration of

RNA was measured using a NanoDrop ND-1000 spectrophotometer (Witec AG, Littau,

Switzerland). The expression of LYVE-1 and Prox1 mRNA was quantified by TaqMan real-

time RT-PCR using the AB 7900 HT fast real-time PCR system (Applied Biosystems, Foster

City, USA). The following probes and primers were used: LYVE-1 forward primer (FP) 5’-

AGCTATGGCTGGGTTGGAGA-3’, reverse primer (RP) 5’ CCCCATTTTTCCCACACTTG-

3’, probe 5’ FAM-TTCGTGGTCATCTCTAGGATTAGCCCAAACC-BKH1-3’; Prox1: FP 5’-

ACAAAAATGGTGGCACGGA-3’, RP 5’-CCTGATGTACTTCGGAGCCTG-3’, probe 5’-

FAM-CCCAGTTTCCAAGCCAGCGGTCTCT-BKH1-3’. Each reaction was normalized for

the expression of ß-actin (FP 5’-TCACCGAGCGCGGCT-3’, RP 5’-

TAATGTCACGCACGATTTCCC-3’, probe 5’-JOE-CAGCTTCACCACCACGGCCGAG-

TAMRA-3’). Student t-test was performed in Office EXCEL. For fluorescence-activated cell

sorting (FACS) analysis, cells were stained with rabbit anti-human LYVE-1 antibody

(ReliaTech, Braunschweig, Germany), anti-rabbit fluorescein isothiocyanate (FITC) or isotype

control antibodies (CALTAG/Invitrogen, Basel, Switzerland), and FACS was performed using a

BD FACSCanto (BectonDickinson, Basel, Switzerland) and the FACSDiva software. Data were

analyzed with Flowjo software (Treestar, Ashland, TN). For immunostains, cells were stained

with human LYVE-1 antibody (ReliaTech) and human Prox1 antibody (kindly donated by Prof.

K.Alitalo, Helsinki, Finland), and corresponding secondary antibodies were labeled with Alexa-

Page 56: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

55

Fluor 488 or 594 (Molecular Probes). Cell nuclei were counterstained with Hoechst

bisbenzimide (Sigma). All experiments were performed three times with comparable results.

4.1.3.3 Immunofluorescence and quantitative analysis of vessel development in EBs

EBs (n=9 per group) were stained with antibodies against mouse LYVE-1 (Angiobio, Del Mar,

CA and R&D Systems), CD31 (BD Bioscience) or Prox1 (kindly provided by Dr. Kari Alitalo),

and corresponding secondary antibodies were labeled with Alexa-Fluor 488 or 594 (Molecular

Probes, Eugene, OR). Cell nuclei were counterstained with Hoechst bisbenzimide (Sigma-

Aldrich). LYVE-1/CD31 stained sections were examined with a Zeiss Axiovert 200M

microscope, and images were captured with a Zeiss AxioCam-MRm (Carl Zeiss; Oberkochen,

Germany). Image acquisition in the individual fluorescent channels was accomplished using

Axio Vision4.4 software (Zeiss). Adobe Photoshop CS3 (Adobe Systems, San Jose, CA) was

used to adjust image brightness and for image overlay. Computer-assisted morphometric vessel

analyses were performed using the IP-LAB software (Scanalytics; Fairfax, VA). The total EB

area was examined and the vessel area and number per EB were determined. The average vessel

area and vessel number per group was then calculated and statistical analysis (unpaired

Student’s t-test) was performed using MS Excel 2003. Prox1/LYVE-1/CD31 positive cell

cluster imaging and quantification was done with a Delta Vision microscope at 20x, and the 3-

dimensional images were captured with a Roper CoolSnap HQ camera and analyzed with

DeltaVision Analysis Software (Applied Precision SoftWoRx, Issaquah, WA) for

deconvolution.

4.1.3.4 Immunohistochemistry of mouse embryo sections

FVB mice (12 to 16 weeks of age; 2 estrous females and 1 male per cage) were allowed to breed

overnight. Females with detectable vaginal plugs on the next morning were determined to be at

Page 57: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

56

day 0 of pregnancy. Pregnant mice were housed individually. The pregnant mice were sacrificed

by CO2 at days 9.5, 10.5 and 11.5 of pregnancy. The embryos were removed by laparotomy. All

embryos were immediately fixed in 4% PFA at 4°C for 48 hours, then dehydrated in ethanol

series, cleared in xylene, and embedded in paraffin wax. Serial cross 10 µm-sections of the

embryos were cut and mounted on glass slides. After they were dewaxed in xylene, sections

were hydrated and processed for immunohistochemistry for LYVE-1 (goat biotinylated anti-

mouse; R&D Systems) and RAR-(rabbit anti-mouse; Santa Cruz Biotechnology). In

additional experiments, RA (Sigma-Aldrich) or Ro 41.5253 (BioMol International) were

dissolved in corn oil right before use; pregnant mice (n=5 per treatment group) were given two

intra-peritoneal injections of 25 mg/kg of body weight of RA or 50 mg/kg body weight of Ro

41-5253 in corn oil on days 8 and 10 of pregnancy. Control mice (n=5) received an equal

volume of corn oil. Mice were sacrificed on day 11.5 of pregnancy. Embryos were embedded in

paraffin wax or frozen in OCT (Sakura FineTek, Torrance, CA). Serial cross sections of the

embryos were cut at a thickness of 10 µm (paraffin) or 15 µm (frozen sections) and were

mounted on glass slides. Paraffin sections were processed for LYVE-1 immunohistochemistry

as described above. The sections were exposed to AEC (3-amino-9-ethylcarbazole) chromogen

that forms a red end-product at the site of the antigen and the reaction was stopped after 1

minute incubation. Frozen sections were fixed in 100% methanol at –20°C for 10 minutes and

then processed for immunofluorescence staining for Prox1 (kindly donated from Prof. K

Alitalo) and CD31 (BD Bioscience), using corresponding secondary antibodies labelled with

Alexa-Fluor 488 or 594 (Molecular Probes, Eugene, OR). Cell nuclei were counterstained with

Hoechst bisbenzimide. The stained sections were examined with a Zeiss Axioskop 2 mot plus.

All images were captured with a Zeiss AxioCam-MRm. The quantification of LYVE-1

expression was performed using Photoshop C3; the number of selected LYVE-1 stained pixels

was determined and their percentage per total pixel number per picture (LYVE-1 area fraction)

was then determined. The jugular lymph sac area quantification was performed using the area

Page 58: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

57

measurement tool of Photoshop C3. The quantification of CD31+/Prox1+ cells was performed

by counting the number of CD31+/Prox1+ cells per picture. Three sections per embryos and

four embryos from four different pregnant mice were analyzed.

4.1.3.5 Xenopus laevis embryos and in situ hybridization

In vitro fertilization, culture and staging of Xenopus laevis embryos (tadpoles) were performed

as previously described (Brandli and Kirschner 1995; Helbling et al. 1999). Embryos were

cultured in 0.1X MMR (0.1 M NaCl, 2 mM KCl, 1 mM MgSO4, 2 mM CaCl2, 5 mM HEPES,

and 0.1 mM EDTA) at 22°C until stage 28. Thereafter, 20 µM RA plus 1 mM cAMP or DMSO

were added to the medium. In addition, tadpoles were also bathed with Ro 41-5253

(concentration range from 100nM to 20µM) but the treatment resulted in lethality within the

first 4 hours of exposure. Embryos were fixed at stage 39 in 4% formalin for 1 hour. Probe

synthesis, whole-mount in situ hybridization and bleaching of embryos were carried out as

described (Helbling et al. 1998; Helbling et al. 1999; Saulnier et al. 2002). For in situ

hybridization, probe synthesis, whole-mount in situ hybridization and bleaching of embryos

were carried out as described (Helbling et al. 1998; Helbling et al. 1999; Saulnier et al. 2002).

Digoxigenin probes were generated from linearized plasmids encoding for LYVE-1, Prox1,

VEGFR-3 and CD31. Digital photographs of stained embryos were taken with an AxioCam

Color camera mounted on a Zeiss Stereo Lumar.V12 stereoscopic microscope.

Page 59: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

58

4.1.4 Results

4.1.4.1 RA promotes lymphatic differentiation of EBs

We used the EB vascular differentiation assay to identify regulators of the early steps of

lymphatic vascular system development (Liersch et al. 2006). We first investigated the effects

of several candidate molecules on the number and size of CD31+/LYVE-1+ vessel-like

structures. The factors were added to EBs, starting at day 14 after initiation, for up to 10 days. In

accordance with previous results (Kreuger et al. 2006; Liersch et al. 2006), VEGF-C promoted

the formation CD31+/LYVE-1+ vascular structures, compared with the vehicle control. The

maximum induction occurred on day 4 of exposure (data not shown), so this was chosen as the

study period for further investigations.

Incubation of EBs with VEGF-C, growth hormone, IGF-1 and IL-7 significantly promoted the

expression of LYVE-1 in CD31+ structures (Figure 4.1.1 A). Incubation of EBs with RA and,

even more potently, with a combination of RA and cAMP, resulted in an enlarged area of

CD31+/LYVE-1+ structures, compared to controls (Figure 4.1.1 A). Only VEGF-C and RA,

with or without cAMP, also significantly promoted the number of CD31+/LYVE-1+ structures

(Figure 4.1.1 B). In contrast, no major effects on the total area or number of CD31+/LYVE-1+

structures were detected after incubation with placental growth factor, hepatocyte growth factor,

IL-3 or the nitric oxide donor S-nitroso-N-acetyl-l,l-penicillamine (SNAP). cAMP did not

enhance VEGF-C effect and incubation with cAMP alone had no effect on EB vasculature

(Figure 4.1.1 A). In agreement with a specific role of retinoic receptors, we found that in

addition to RA, 13-cis-retinoic acid (+cAMP) and the synthetic retinoid analogue TTBNP (+

cAMP) also promoted significant formation of LYVE-1+ /CD31+ area (Figure 4.1.1 C).

Incubation of EBs with RA significantly increased the EB area covered by CD31+/LYVE-1+

structures (Figure 4.1.2 D-F) and, in agreement with the documented synergistic effect of RA

Page 60: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

59

and cAMP in other systems (Lang et al. 1989; Drab et al. 1997; Hatzopoulos et al. 1998), the

combination of RA and cAMP for 4 days resulted in LYVE-1 expression by most of the CD31+

endothelial cells (78.6%+14%; Figure 4.1.2 G–L’), compared to 27%+14% in control EBs

(Figure 4.1.2 A–C). A quantitative analysis revealed that the RA+cAMP treatment significantly

increased the total CD31+ area and the CD31+/LYVE-1+ area, but not the CD31+/LYVE-1-

area, as compared with controls (Figure 4.1.2 M), thus excluding the possibility that the

observed increase of LYVE-1 expression might be a secondary effect due to an overall

increased amount of CD31-positive vascular structures.

Page 61: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

60

Figure 4.1.1 The in vitro mouse embryoid body (EB) assay reveals that retinoic acid (RA) and cAMP induce

LYVE-1 expression. Mouse EBs were cultured for 14 days and were then incubated with compounds for 4 days.

10 µM all-trans-RA (RA) and RA+0.5 mM cAMP (RA+cAMP) increased the LYVE-1+ area among CD31+

vessel-like structures (A). VEGF-C, IGF-1, GH and IL-7 also showed a significant induction of LYVE-1

expression (A). 10 µM all-trans-RA + cAMP and VEGF-C induced the formation of several distinct LYVE1+

lymphatic vessel-like structures (B). 13-cis-RA + cAMP and the synthetic retinoid TTBNP (+ cAMP) increased the

area of LYVE-1 expression (C). Exposure of EBs to Ro 41-5253, an inhibitor of RAR-α, in combination with

RA+cAMP inhibited the expression of LYVE-1 in CD31+ vessel-like structures. H89, an inhibitor of PKA, also

inhibited the effects of RA + cAMP. Ro 41-5253 did not significantly inhibit the VEGF-C effect. EBs exposed to

the inhibitors alone expressed the same levels of LYVE-1 as controls. Data are expressed as mean values + SEM

(n= 5), *p<0.05; **p<0.01;*** p<0.001.

Page 62: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

61

Figure 4.1.2 Incubation of EBs with RA and cAMP induces LYVE-1 expression in CD31+ vessel-like

structures. Differential immunofluorescence analysis of control EBs for expression of CD31 (red) and LYVE-1

(green) showed the formation of CD31+/LYVE-1- blood vessel-like structures whereas only a few endothelial cells

expressed LYVE-1 (A-C). Incubation with RA induced LYVE-1 expression in the CD31+ structures (D-F). RA in

combination with cAMP (RA+cAMP) further increased the CD31+/LYVE-1+ area (G-I, J-L’ and M). The total

CD31+ and the CD31+/LYVE-1+ area, but not the CD31+/LYVE-1- area was increased (M). Scale bars=100 µm;

data are expressed as mean values + SEM (n=9).

Page 63: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

62

Importantly, incubation of EBs with RA and cAMP for up to 4 days significantly increased the

number of Prox1+/LYVE-1+/CD31+ cell clusters formed (Figure 4.1.3 E-J ; Figure 4.1.4, E-H),

whereas only a few individual Prox1+/LYVE-1+/CD31+ cells, but no cell clusters, were

detected in control EBs (Figure 4.1.3 A-D and J plus Figure 4.1.4 A-D).

Figure 4.1.3 Incubation of EBs with RA and cAMP increases formation of Prox1+/LYVE-1+/CD31+ cell

clusters. Double immunofluorescence analysis revealed that incubation with RA and cAMP induced expression of

Prox1 (green; F, G, H, I) in CD31+ (red; E, G, I) and in LYVE-1+ (Figure 4.1.4 E-H) endothelial cell clusters,

compared with control EBs where only a few Prox1+ (B, C and D) and CD31+ (A, C) cells were visible. Cell

nuclei are stained blue (D, H). Pictures were obtained after 2 days of exposure to RA and cAMP. Scale bars=50

µm. Quantitative analysis revealed that RA and cAMP exposure increased the number of CD31+/Prox1+ cell

clusters in EBs compared with negative controls, beginning at 1 day after treatment initiation (J). Data are

expressed as mean values + SEM (n= 5), *p< 0.05; **p<0 .01; ***p<0 .001.

Page 64: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

63

Figure 4.1.4 Prox1 positive endothelial cell clusters in

the EBs are also LYVE-1 positive. Double

immunofluorescent staining for LYVE-1 (red) and Prox1

(green) showed that EB treatment with RA+cAMP induced

expression of Prox1 (F, G) in LYVE-1+ (E, G) endothelial

cell clusters as compared with control (A-D) Cell nuclei:

blue (D, H). Scale bars: 20 µm.

We next investigated whether the effects of RA and cAMP on the formation of lymphatic

vessel-like structures was inhibited by the RAR-–specific antagonist Ro 41-5253. Incubation

of EBs with Ro 41-5253 alone did not affect the formation of CD31+/LYVE-1+ or

CD31+/LYVE-1+/Prox1+ structures, whereas Ro 41-5253 potently inhibited the induction of

lymphatic vessel-like structures by RA and cAMP (Figure 4.1.1 C and Table 4.1.1).

Page 65: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

64

Table 4.1.1 Day 16 Day 18

#EBs #Clusters #EBs #Clusters

Control 0/9 0 0/9 0

10 µM RA 0/9 0 0/9 0

10 µM RA+0.5 mM cAMP 9/9 3+1 9/9 1+0

10 µM RA+0.5 mM cAMP+10 µM Ro 41-5253 0/9 0 0/9 0

10 µM RA+0.5 mM cAMP+10 µM H89 0/9 0 0/9 0

0.5 mM cAMP 0/9 0 0/9 0

10 µM Ro 41-5253 0/9 0 0/9 0

10 µM H89 0/9 0 0/9 0

Table 4.1.1 Blockade of RAR- or of PKA inhibits the induction of Prox1+ cell clusters by RA and cAMP.

Four days of treatment with RA and cAMP induced the formation of 3+1 Prox1+/LYVE-1+/CD31+ cell clusters

per EB. These clusters were found in 9 out of 9 EBs analyzed. Ro 41-5253 (RAR-inhibitor) and H89 (PKA

inhibitor) completely prevented the induction of Prox1 endothelial cell clusters by RA and cAMP. No induction

was seen after incubation with RA alone, cAMP alone or with the inhibitors alone. #EB: number of analyzed EBs;

#Clusters: average number of CD31+/LYVE-1+/Prox1+ cell clusters per EB + SD.

The cAMP-dependent PKA inhibitor H89 also completely blocked the induction of

CD31+/LYVE-1+/Prox1+ structures by RA and cAMP; incubation of EBs with H89 alone had

no effects (Figure 4.1.1 C and Table 4.1.1). Together, these findings indicate a sufficient role of

the RAR–α and cAMP–PKA pathway in promoting the retinoid effects on lymphatic

differentiation in the mouse EB assay.

4.1.4.2 In vivo effects of RA in developing mouse embryos

We investigated whether RA could affect in vivo development of the mouse lymphatic vascular

system. Immunohistochemical analyses were used to determine whether RA receptors are

expressed by endothelial cells of the cardinal vein of mouse embryos at ED 9.5–11.5, when

Page 66: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

65

expression of LYVE-1 and Prox1 is first observed. We found that RAR- was expressed

(Figure 4.1.5, B’, arrowheads) by the endothelial cells of the LYVE-1+ cardinal vein (Figure

4.1.5 A’, arrowheads) and by the developing lymph sacs at ED 11.5. In fact, RAR- was

expressed on/nearby the cardinal veins by EDs 10.5 (Figure 4.1.5 C) and 9.5 (Figure 4.1.5 D),

time-points at which the jugular lymph sacs had not yet formed.

Figure 4.1.5 RAR- is expressed by endothelial cells of the cardinal veins of mouse embryos.

Immunohistochemical analysis of mouse embryos for LYVE-1 (red; A-A’: ED 11.5) and RAR- (B, B’: ED11.5;

C: ED10.5; D: ED9.5) revealed that RAR- (B, B’, arrowheads) is expressed by/near the endothelial cells of the

LYVE-1+ (A, A’, arrowheads) cardinal veins and forming jugular lymph sacs of ED 11.5 as well as in/near the

cardinal veins of ED 10.5 (C, arrowheads) and ED 9.5 (D, arrowheads) mouse embryos. Two serial sections are

shown in panels A and B. Scale bars: A, B: 100 µm; A’, B’, C, D: 20 µm. Cell nuclei were counterstained with

hematoxylin. a, dorsal aorta; jls, jugular lymph sac; nt, neural tube; v, cardinal vein.

Page 67: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

66

Based on the observed expression pattern of RAR- during lymphatic development, we

investigated whether RA also induced in vivo expression of LYVE-1 and Prox1. To this end, we

injected RA intraperitoneally into pregnant mice, to expose the developing embryos to an in

utero excess of RA. RA (25 mg/kg of weight) was injected on days 8 and 10 of pregnancy. ED

8 was chosen as the first injection time-point to ensure RA exposure before LYVE-1 and Prox1

were expressed by cardinal vein endothelium (at ED 9). ED10 was chosen for the second

injection to ensure that lymphatic-committed endothelial cells were exposed to excess RA as

they were budding from the cardinal veins.

At ED 11.5, when the first jugular lymph sacs are visible, immunohistochemistry with a short 1

min colorimetric reaction revealed strong LYVE-1 expression on the cardinal vein endothelial

cells of embryos that were exposed to RA in utero (Figure 4.1.6 C, C’ and D) but not of control

embryos (Figure 4.1.6 A, A’ and B). A quantitative analysis confirmed that LYVE-1 expression

was significantly upregulated upon treatment (Figure 4.1.6 G). We also observed an increase in

the size of the primary jugular lymph sacs in embryos exposed to RA (Figure 4.1.6 H; p=0.19).

In contrast, in utero exposure of the embryos to Ro 41-5253, an inhibitor of RAR-α, led to a

decrease in LYVE-1 expression by the ECs of the cardinal vein and of the jugular lymph sac

(Figure 4.1.6 E-F and G), as compared to control embryos (Figure 4.1.6 A, A’, B, G). The

lymph sac area was not affected by Ro 41-5253 (Figure 4.1.6 H). Importantly, differential

immunofluorescence analyses of CD31 and Prox1 expression revealed that embryonic exposure

to RA increased the number of Prox1+ endothelial cells in the cardinal veins and of sprouting

Prox1+ cells that form the lymph sacs (Figure 4.1.7 D-D’’’, E, and F), compared with control

embryos (Figure 4.1.7 A-A’’’, B and C). A quantitative analysis confirmed that the number of

CD31+/Prox1+ cells in the jugular area of the embryos significantly increased upon RA

treatment (Figure 4.1.7 I), whereas injection of Ro 41-5253 slightly decreased the number of

Page 68: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

67

CD31+/Prox1+ cells (Figure 4.1.7 G-G’’’, H and I) as compared to controls (Figure 4.1.7 A-

A’’’, B, C and I).

Figure 4.1.6 In utero exposure of mouse embryos to excess of RA upregulates endothelial LYVE-1 expression

in the anterior cardinal veins and jugular lymph sacs. Immunohistochemical analysis of paraffin sections from

ED 11.5 mouse embryos for LYVE-1 (red) revealed that the endothelial cells of the anterior cardinal veins and of

the forming lymph sacs upregulated the expression of LYVE-1 after in utero exposure to an excess of RA (C, C’, D

and G), as compared with control embryos (A, A’, arrowheads, B and G). Treatment with the RAR antagonist Ro

41-5253 decreased LYVE-1 expression (E, E’, F and G). The jugular lymph sac area was enlarged after RA

treatment as compared to controls (H). A' - B, C' - D and E' - F are sections of embryos from two independent

pregnant mice each. Scale bars: A, C, E, 100 µm; A’-F, 50 µm. Jls, jugular lymph sac; nt, neural tube; v, cardinal

vein; data are expressed as mean values + SD (n=4) **p<0 .01.

Page 69: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

68

Figure 4.1.7 In utero exposure of mouse embryos to excess of RA increases the number of Prox1-positive

lymphatic progenitor cells in the cardinal veins and jugular lymph sac forming areas. Double-

immunofluorescence analysis of ED 11.5 mouse embryos exposed to excess RA (D-D’’’, E and F) or vehicle (A-

A’’’, B and C) for CD31 (green) and Prox1 (red) revealed that RA increased the number of Prox1+ endothelial

cells in the anterior cardinal veins and in the area of the jugular lymph sacs as compared with control (I). Ro 41-

5253 treatment (G-G’’’ and H) slightly reduced the number of Prox1+/CD31+ cells in the jugular area as compared

with control (I). A-B-C, D-E-F and G-H show representative sections of embryos from independent treated

Page 70: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

69

pregnant mice, respectively. Cell nuclei: blue (A’, D’, G’). Scale bars: A, A’,D, D’,G, G’: 100 µm; A’’-F: 50 µm.

a, dorsal aorta; jls, jugular lymph sac; nt, neural tube; v, cardinal vein; data are expressed as mean values + SD

(n=4) ***p<0 .001.

The increase in CD31+/Prox1+ cells in the cardinal veins was not due to enhanced cell

proliferation, as revealed by the results of phosphohistone 3 stains (data not shown). ED 11.5

embryos, at gross examination, showed a slight increase in blood presence in the extra-

embryonic tissues, a lower amount of blood in the heart region as well as a lower heart beat

frequency. The treatment also caused a mild reduction of the caudal length as previously shown

(Chan et al. 2002) (data not shown).

4.1.4.3 RA upregulates expression of LYVE-1 and VEGFR-3 in the vasculature of

Xenopus laevis embryos

Due to their small size, transparency and easy maintenance, Xenopus laevis embryos are a

useful animal model for studying development (Dawid and Sargent 1988), particularly that of

the lymphatic vasculature (Ny et al. 2005). Xenopus embryos were incubated with RA and

cAMP from stage 28 on, when the Prox1 expression is first detected on the vasculature, until

stage 39, when lymphatic endothelial cells start to sprout from the lymph heart and the cardinal

vein to form the first lymphatic vessels. In situ hybridization analyses revealed that exposure of

the embryos to RA increased expression of VEGFR-3 (Figure 4.1.8 G, H and H’) and LYVE-1

(Figure 4.1.8 E, F and F’) throughout the developing vasculature, including the intersomitic

veins (arrowheads in Figure 4.1.8 F and H, H’), the cardinal veins (arrows in Figure 4.1.8 F and

H, H’) and the vitelline network (asterisks in Figure 4.1.8 H), indicative of enhanced lymphatic

competence. Expression of CD31 was downregulated throughout the entire vasculature (Figure

4.1.8 A, B, B’); this was an important observation because CD31 expression is reduced on

lymphatic vessels, compared to blood vessels, during normal development. Prox1+ cells were

Page 71: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

70

found in the anterior and posterior cardinal veins, the tip of the tail, the lymph heart and the first

sprouts from the lymph heart in both the RA-exposed and control embryos (Figure 4.1.8 C, D,

D’). A quantitative analysis of the VEGFR-3 expression revealed a significant increase in the

number of VEGFR-3+ sprouts that developed from the anterior cardinal vein and the lymph

heart following exposure to the combination of RA and cAMP, compared with controls

(p=0.002; Figure 4.1.8 I). Only 16%+5% of the control embryos had > 5 VEGFR-3+ sprouts,

compared to 54%+5% of RA-exposed embryos. Moreover, in the embryos exposed to RA,

VEGFR-3+ sprouts were longer than those of the control group (Figure 4.1.8 G, H, H’).

Figure 4.1.8 Incubation of Xenopus laevis embryos with excess of RA downregulates CD31 and upregulates

the lymphatic markers LYVE-1 and VEGFR-3 in the developing vasculature. Xenopus laevis tadpoles were

bathed in RA in combination with cAMP (B, B’, D, D’, F, F’and H, H’) or in DMSO (A, C, E and G) from

embryonic stages 28 to 39. In situ hybridization (blue) for CD31 (A, B, B’), Prox1 (C, D, D’), LYVE-1 (E, F, F’)

or VEGFR-3 (G, H, H’) revealed that RA and cAMP reduced CD31 expression (B, B’) in the developing

vasculature, including intersomitic veins (isv), the cardinal vein (v), and the vitelline network (vn), compared with

controls (A). No effects on Prox1 expression and/or distribution in the lymph heart (lh), lymphatic sprouts (ls) or in

the lymphatic endothelial cells of venous origin (lv) were observed (C, D, D’). In contrast, LYVE-1 expression was

Page 72: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

71

upregulated in the intersomitic veins and in the cardinal vein (F, arrowheads, F’) compared with controls (E). RA

incubation also upregulated VEGFR-3 expression in the cardinal vein and vitelline network (H, arrowheads and

asterisk respectively and H’) compared with control embryos (G, arrow heads). Scale bars: 50 µm; Scale bars of

insert pictures: 5 µm. isv, inter somatic veins; lh, lymph heart; ls, lymphatic sprouts; lv, lymphatic endothelial cells

of venous origin; v, cardinal vein; vn, vitelline network. (I) Quantitative analysis of the number of VEGFR-3+

sprouts from the anterior cardinal vein following exposure to DMSO (vehicle) or the combination of RA and

cAMP. Data are expressed as mean + SEM (n=150 per treatment group).

4.1.4.4 Human umbilical vein endothelial cells upregulate LYVE-1 expression upon

RA+cAMP treatment

We investigated whether RA in combination with cAMP could induce the expression of LYVE-

1 and Prox1 in human umbilical vein endothelial cells (HUVECs) in vitro.

Exposure of HUVECs to RA and cAMP for 12h, led to an upregulation of LYVE-1 mRNA

expression as shown by real-time RT-PCR (>2-fold; p=0.042; Figure 4.1.9 A). Expression of

Prox1 mRNA was unchanged (data not shown). Furthermore, the treatment resulted in an

upregulation of LYVE-1 at the protein level as shown by FACS analysis (Figure 4.1.9 B).

Double immunofluorescence stains for LYVE-1 and Prox1 of HUVECs treated or not with

RA+cAMP showed an upregulation of LYVE-1 but not Prox1 protein expression after treatment

(Figure 4.1.9 C).

These observations, showing that RA induces expression of LYVE-1 in human endothelial

cells, suggest that RA’s effects observed in the EBs in vitro and on embryos in vivo might be

directly mediated by endothelial cells.

Page 73: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

72

Figure 4.1.9 HUVECs upregulate LYVE-1 expression after RA+cAMP treatment. (A) Exposure of HUVECs

to RA and cAMP for 12h, led to an upregulation of LYVE-1 mRNA expression as shown by real-time RT-PCR;

data are expressed as mean fold change + SEM (n=3). (B)Treatment resulted in an upregulation of LYVE-1 at the

protein level as shown by FACS analysis. Red profile indicates negative controls, whereas LYVE-1 antibody

reactivity is plotted as blue profile. X-axis represents the fluorescence intensity on a logarithmic scale whereas the

Y-axis shows number of events normalized to the highest cell count; one representative experiment out of three is

depicted; (C) Double immunofluorescence stains for LYVE-1 (red) and Prox1 (green) of HUVECs treated or not

with RA+cAMP showed an upregulation of LYVE-1 but not Prox1 protein expression after treatment; cell nuclei

blue; scale bar 20 µm.

Page 74: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

73

4.1.5 Discussion

This is the first demonstration that all-trans-RA is involved in the earliest steps of lymphatic

vascular development, namely the acquisition of lymphatic competence and commitment by

endothelial cells of the embryonic cardinal vein; RA appears to do so by upregulating the

lymphatic markers LYVE-1 and Prox1.

Our studies with the mouse EB vascular differentiation assay found that VEGF-C, growth

hormone, IGF-1 and IL-7 increased the area of CD31+/ LYVE-1+ vessel-like structures in the

EB assay; these findings are in agreement with the reported lymphangiogenic activity of growth

hormone (Banziger-Tobler et al. 2008) and IGF-1 (Bjorndahl et al. 2005) and the lymphatic

reprogramming activity of IL-7 in cultured endothelial cells (Al-Rawi et al. 2005). Most

interestingly, however, we found that RA, alone and combination with cAMP, potently up-

regulated LYVE-1 expression in CD31+ vascular structures. The LYVE-1+, lymphatically

competent endothelial cells emerged from the pre-existing CD31+ blood vascular endothelium,

supporting the centrifugal theory of lymphatic vasculature development from pre-existing

embryonic veins originally proposed by Sabin in 1902 (Sabin 1902). However, the combination

of RA and cAMP also induced the formation of rare CD31+/LYVE-1+/Prox1+ cell clusters,

independently from the CD31+ vessel-like networks. These findings indicate that mesenchymal

progenitor cells might differentiate directly into lymphatically competent endothelial cells; this

is in agreement with the centripetal theory of mesenchyme-derived lymphatic progenitors

proposed by Huntington in 1910 (Huntington and McClure 1910). The mouse EB vascular

differentiation assay represents a suitable model for lymphatic vasculature development in vivo,

since lymphatic vessels in mice appear to be predominantly derived from pre-existing

embryonic veins, as suggested by recent lineage-tracing studies (Srinivasan et al. 2007),

although there might be a contribution by mesenchyme-derived progenitor cells (Wilting et al.

2003). EB cells appear to be more responsive to the induction of lymphatic commitment than

Page 75: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

74

differentiated endothelial cells, since there was no induction of Prox1 by RA and cAMP in

cultured human umbilical vein endothelial cells (HUVECs). The absence of Prox1 induction in

HUVECs, in a 2-dimensional culture system, might indicate the need for additional inducing

factors derived from the microenvironment that are present in the EBs.

RA binds to nuclear retinoic acid receptors (RARs), which after forming heterodimers with

retinoid X receptors (RXRs), activate histone acetylation and gene transcription. Our finding

that Ro 41-5253 inhibited the induction of lymphatic vascular structures by RA indicated that

the RA effect was mediated via RAR-. Ro 41-5253 antagonizes the transactivation of RARs

by RA, with a high affinity for RAR-; 50- to 100-fold higher concentrations of Ro 41-5253 are

required to reduce the activation of RAR- and 1000-fold higher concentrations are required to

inhibit RAR-Apfel et al. 1992). The potentiation of RA’s effects by cAMP and the inhibition

by H89, a cAMP-dependent PKA inhibitor, indicate that RA’s effects are mediated by a

pathway that includes cAMP and PKA (Gaillard et al. 2006). Indeed, it has been found that RA

and cAMP have synergistic effects in several differentiation processes including neural (Lang et

al. 1989), smooth muscle (Drab et al. 1997) and endothelial progenitor cell (Hatzopoulos et al.

1998) differentiation, although the precise mechanisms of RA and cAMP interaction remain to

be established. This concept is supported by our in silico analyses; the AliBaba

(http://www.gene-regulation.com/pub/programs.html), PROMO (http://alggen.lsi.upc.es/cgi-

bin/promo_v3/promo/promoinit.cgi?dirDB=TF_8.3) and TESS (http://www.cbil.upenn.edu/cgi-

bin/tess/tess) software indicates that RXRs and RARs, as well as cAMP response element-

binding proteins (CRE-BP), bind to the promoter regions of the mouse LYVE-1 and Prox1

genes (data not shown).

The role of retinoid signalling in the mediation of lymphatic competence is supported by

immunohistochemical analyses showing that RAR- is strongly expressed by the endothelial

Page 76: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

75

cells of the embryonic cardinal veins at ED 9.5, when lymphatic competence and commitment

are first detected and at EDs 10.5 and 11.5, when LEC progenitors bud off from the cardinal

vein (Wigle et al. 2002; Oliver 2004). The finding that at EDs 9.5 and 10.5, RAR- was also

expressed by some cells in clusters near cardinal veins, raises the possibility that surrounding

cells might contribute, by indirect effects, to the induction of lymphatic competence by RA.

However, our observation that incubation of HUVECs with RA increased LYVE-1 mRNA

expression and also increased LYVE-1 protein levels, suggests that RA’s effects are directly

mediated by endothelial cells.

To investigate whether RA also promotes lymphatic competence and commitment in vivo, we

used established models of Xenopus and mouse lymphatic development. Previous studies have

shown that excess or lack of RA during early embryogenesis causes malformations (Lammer et

al. 1986; Radhika et al. 2002), through mispatterning of the embryonic tissue layers and the

anterior–posterior body axis. Several RAR knockout mice have been created (Clagett-Dame and

DeLuca 2002). Together, RAR deficiency and vitamin A deprivation lead to a plethora of

embryonic defects, including those of the eye, respiratory tract, heart, kidney, genital tract,

bones and limbs, as well as axial skeletal and neural malformations (Lohnes et al. 1995).

Importantly, however, vitamin A deficiency is also associated with cardiovascular problems and

enlargement of the anterior cardinal veins in rat embryos (White et al. 1998; White et al. 2000).

Zebrafish, that express mutant forms of the RA-metabolizing enzyme Cyp26A1, also display

patterning defects in multiple organs, including the common cardinal vein, pectoral fin, tail,

hindbrain, and spinal cord. Because of the multitude of developmental defects caused by RA

deficiency or exposure (Kochhar 1967; Moro Balbas et al. 1993; Ozeki and Shirai 1998;

Quemelo et al. 2007; Duester 2008), we investigated the in vivo effects of RA exposure over

only short time periods and during the earliest phases of lymphatic development.

Importantly, in mice increased levels of RA in utero upregulated LYVE-1 expression in the

endothelial cells of the anterior part of the cardinal veins and of the forming lymph sacs (which

Page 77: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

76

also slightly increased in size). An increased number of CD31+/Prox1+ endothelial cells were

detected in the cardinal veins and the developing lymph sacs. In contrast, in utero injection of

Ro 41-5253, inhibiting RAR-α signalling (Apfel et al. 1992), reduced LYVE-1 expression and

decreased the number of CD31+/Prox1+ cells in the jugular area of the ED11.5 embryos.

However, the extent of inhibition was rather mild and the quantitative evaluation did not reach a

significance level of 0.05, in part due to the rather high variability among the different embryos.

Other possible explanations for these findings are 1) a potential compensatory role of RAR-β

and/or –γ (Manshouri et al. 1997), 2) non-canonical activity of RA via CREB or ERK (Canon et

al. 2004), and 3) an incomplete blockade of RAR-α activity by the two injections of Ro 41-

5253.

Exposure of stage 28–39 Xenopus embryos to RA down-regulated CD31 expression but

enhanced expression of VEGFR-3 and LYVE-1 in the developing vasculature, mainly in the

intersomitic veins, the cardinal vein and the vitelline vein network. Because incubation of

Xenopus embryos with different concentrations of Ro 41-5253 induced lethality of stage 28

tadpoles (data not shown), the vascular effects of specific blockade of RAR-α could not be

evaluated in the Xenopus model. In addition, morpholino knockdown experiments would not

represent a suitable loss-of-function approach since it has been shown that knockdown of either

RAR- α1 or -α2 (or both) leads to a severe phenotype in Xenopus embryos, mainly regarding the

head formation, already at the early stage 20 (Koide et al. 2001) (the first Prox1 expression in

Xenopus is at stage 28).

All-trans-retinoic acid and its derivatives exert many different and often contradictory effects on

cells, depending on context, cell type, and other variables; its role in angiogenesis, for example,

remains so far controversial. It is has been found that RA induces in vitro tube formation of

HUVECs (Saito et al. 2007) via a possible paracrine effect by inducing endogenous VEGF-A

and FGF2 production, and angiogenesis in bovine aortic endothelial cells in vitro (Gaetano et al.

Page 78: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

77

2001). On the other hand, RA caused endothelial cells to become refractory to stimulation by

either tumor-conditioned media or various angiogenic factors without interfering with cell

proliferation (Lingen et al. 1996). In addition, there is evidence that the anti-tumor activity of

retinoids involves inhibition of tumor-induced angiogenesis (Oikawa et al. 1989; Majewski et

al. 1995; Lingen et al. 1996).

Interactions between retinoid signalling and Prox1 expression have been described during other

aspects of development. RA modulates Prox1 expression in the dorsal endoderm of mouse

embryos (Burke and Oliver 2002) and promotes neuronal progenitor cell differentiation into

retinal (Zhao et al. 2002) or cochlear cells (Bermingham-McDonogh et al. 2006), which express

Prox1 (Tomarev et al. 1996; Wigle et al. 2002; Dyer 2003; Bermingham-McDonogh et al.

2006). Development of the murine liver also appears to be RA-dependent (Ogura et al. 2004)

and Prox1 is a marker for rat and mouse hepatocytes (Burke and Oliver 2002; Dudas et al.

2006). Moreover, RA, Prox1 and RALDH2 all have important roles in pancreas development

(Burke and Oliver 2002; Molotkov et al. 2005).

Now, retinoid signalling is shown to mediate the earliest steps of lymphatic vasculature

development-the acquisition of lymphatic competence and commitment via expression of

LYVE-1 and Prox1, respectively. It will be important to determine whether retinoids also

modulate post-natal lymphangiogenesis, such as the growth of new lymphatic vessels during

tissue regeneration and cancer progression (Cueni and Detmar 2008).

Page 79: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

78

4.2 Activation of the epidermal growth factor receptor

promotes lymphangiogenesis in vitro and in vivo

4.2.1 Abstract

The lymphatic vascular system regulates tissue fluid homeostasis and has an important role in

immune surveillance, inflammation and cancer metastasis. However, the molecular

mechanisms involved in lymphangiogenesis remain poorly characterized. We used our

previously established mouse embryonic stem cell-derived embryoid body vascular

differentiation assay to investigate the effects of candidate small molecules on blood and

lymphatic vessel formation. We found that inhibition of the epidermal growth factor (EGF)

receptor (EGFR) inhibited the formation of lymphatic vessel-like structures. In vitro studies

with human dermal lymphatic endothelial cells (LECs), that were found to express EGFR,

revealed that EGF promotes lymphatic vessel formation; this effect was inhibited by

incubation with an EGFR blocking antibody and with small molecules that target the EGFR or

its associated tyrosine kinase. Incorporation of EGF into a matrigel plug assay revealed that

EGF also promotes lymphangiogenesis in vivo. Moreover, transgenic mice with skin-specific

overexpression of amphiregulin, which also activates EGFR/ErbB2, showed an enhanced

density of lymphatic vessels in the skin. Together, our findings reveal, for the first time, a role

for EGFR activation in lymphangiogenesis and suggest that specific EGFR antagonists might

be used to inhibit pathological lymphangiogenesis.

Page 80: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

79

4.2.2 Introduction

The lymphatic vascular system plays an essential role in the maintenance of normal fluid

homeostasis as well as in several pathological conditions, including inflammation and tumor

metastasis. Identification of lymphatic endothelium-specific markers, such as podoplanin and

the lymphatic vessel endothelial hyaluronan receptor-1 (LYVE-1), and studies of lymphatic

system development in a range of genetic mouse models have significantly increased our

understanding of how lymphatic endothelial cell (LEC) differentiation, growth and function

are regulated (Cueni and Detmar 2006). Vascular endothelial growth factor-C (VEGF-C) is,

thus far, the best characterized lymphangiogenic factor that activates VEGF receptor

(VEGFR)-3. Under normal conditions, VEGFR-3 is specifically expressed by LECs but not

by blood vascular endothelial cells (BECs). Activation of VEGFR-3 promotes LEC

proliferation and migration in vitro (Goldman et al. 2007), and lymphatic vessel formation in

vivo (Veikkola et al. 2001). In addition, VEGF-A has also been shown to stimulate

lymphangiogenesis (Hong et al. 2004; Bjorndahl et al. 2005; Hirakawa et al. 2005).

Furthermore, additional growth factors, including fibroblast growth factor-2, hepatocyte

growth factor, angiopoietin-1 and -2, and platelet-derived growth factor, have been recently

shown to promote lymphangiogenic processes (Cueni and Detmar 2008). Because of the

emerging role of the lymphatic vascular system in human disease, including cancer metastasis,

chronic inflammation, organ transplant rejection and hypertension, the identification of

molecular mediators and/or inhibitors of lymphangiogenesis is of immense interest.

Based on the results of a recent chemical library screening for compounds that interfere with

vascular development in Xenopus laevis tadpoles (Kalin et al. 2009), we selected several

compounds to investigate for their possible anti-lymphangiogenic activity in our previously

established mouse embryonic stem cell-derived embryoid body (EBs) vascular differentiation

Page 81: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

80

assay (Liersch et al. 2006). In particular, we investigated the effects of calcium channel

inhibitors and of inhibitors of the EGFR/ErbB2 tyrosine kinase, as well as an inhibitor of

VEGF receptor tyrosine kinases. GW2974, a small molecule that targets the EGFR/ErbB2

tyrosine kinase, strongly inhibited the formation of CD31+/LYVE-1+ lymphatic vessel-like

structures in EBs. Conversely, its ligand EGF promoted the vessel formation in the EBs and

also the in vitro migration and tube formation of human lymphatic endothelial cells (LECs),

that we found to express the EGFR in vitro and in vivo. This effect was inhibited by

incubation with an EGFR blocking antibody and with small molecules that target the EGFR or

its associated tyrosine kinase. Incorporation of EGF into a matrigel plug assay revealed that

EGF also promotes lymphangiogenesis in vivo. Moreover, transgenic mice with skin-specific

overexpression of amphiregulin, which also activates EGFR/ErbB2, showed an enhanced

density of lymphatic vessels in the skin. Together, our findings reveal, for the first time, a role

for EGFR activation in lymphangiogenesis and suggest that specific EGFR antagonists might

be used to inhibit pathological lymphangiogenesis.

4.2.3 Materials and methods

4.2.3.1 Mouse embryonic stem cell culture, establishment and treatment of EBs

Murine C57BL/6x129SvEv derived embryonic stem cells (mES cells; passage 3-12; kindly

provided by N. Gale, Regeneron Pharmaceuticals, Tarrytown, NY), were cultured on

mitotically inactivated primary mouse embryonic fibroblasts (PMEFs, passage 2-5, Institute of

Laboratory Animal Science, University of Zurich, Switzerland) in Dulbecco’s modified Eagle

medium (Gibco, Eggenstein, Germany), supplemented with 18% fetal bovine serum (Gibco),

100 nM sodium pyruvate (Sigma-Aldrich, Buchs, Switzerland), MEM vitamins, 2 mM L-

glutamine, streptomycin and penicillin (all from Gibco), 10 mM 2-mercaptoethanol and 2000

U/ml recombinant leukemia inhibitory factor (LIF; Chemicon International, Temecula, CA).

Page 82: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

81

PMEFs and LIF were removed and mES cells were transferred to suspension culture for

embryoid body (EB) formation as described (Kurosawa 2007). After 3 or 4 days, EBs of the

same size were transferred into 12-well dishes (BD Bioscience, San Diego, CA) and cultured

for 14 days without LIF. Then, EBs were incubated or not with the following factors for 4

days: 100 ng/ml human recombinant epidermal growth factor (EGF, BD Biosciences); 10 µM

all-trans-retinoic acid (RA; Sigma-Aldrich); 0.5 mM 3',5'-cyclic monophosphate (cAMP;

Fluka, Buchs, Switzerland); 200 ng/ml recombinant human VEGF-C (R&D Systems,

Minneapolis, MN) in combination or not with the following substances (all from Sigma-

Aldrich) that were added at 10 µM concentrations: plendil, 4-(2,3-dichlorophenyl)-1,4-

dihydro-2,6-dimethyl-3,5-pyridinecarboxylic acid ethyl methyl ester (felodipine); YC-93, 1,4-

dihydro-2,6-dimethyl-4-(3-nitrophenyl)methyl-2-[methyl(phenylmethyl)amino]-3,5-

pyridinedicarboxylic acid ethyl ester hydrochloride (nicardipine); 4′,5,7-trihydroxyisoflavone,

5,7-dihydroxy-3-(4-hydroxyphenyl)-4H-1-benzopyran-4-one (genistein); N4-(1-benzyl-1H-

indazol-5-yl)-N6,N6-dimethyl-pyrido[3,4-d]pyrimidine-4,6-diamine (GW2974). Medium only

and dimethyl sulfoxide (DMSO) were used as negative controls. EBs were fixed in 20°C cold

methanol for 10 minutes.

4.2.3.2 Immunofluorescence analysis of vessel development in EBs

EBs (n=9 per group) were stained with antibodies against mouse LYVE-1 (Angiobio, Del

Mar, CA and R&D Systems), CD31 (BD Bioscience), and corresponding secondary

antibodies labelled with Alexa-Fluor 488 or 594 (Molecular Probes, Eugene, OR). Cell nuclei

were counterstained with Hoechst bisbenzimide (Sigma-Aldrich). CD31/LYVE-1 stained

sections were examined with a Zeiss Axiovert 200M microscope, and images were captured

with a Zeiss AxioCam-MRm (Carl Zeiss; Oberkochen, Germany). Image acquisition in the

individual fluorescent channels was accomplished using Axio Vision4.4 software (Zeiss).

Page 83: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

82

Adobe Photoshop CS3 (Adobe Systems, San Jose, CA) was used for image overlay.

Computer-assisted morphometric vessel analyses were performed using the IP-LAB software

(Scanalytics; Fairfax, VA). The total EB area was examined, and the vessel area and number

per EB were determined. The average vessel area and vessel number per treatment group was

then calculated and statistical analysis (unpaired Student’s t-test) was performed using MS

Excel 2003.

4.2.3.3 Proliferation, migration and tube formation of human lymphatic endothelial cells

Human dermal microvascular lymphatic endothelial cells (LECs) were isolated from neonatal

human foreskins by immunomagnetic purification as described (Kajiya et al. 2005). Cells were

cultured in endothelial basal medium (Cambrex, Verviers, Belgium) supplemented with 20%

fetal bovine serum (Gibco, Paisley, UK), antibiotic antimycotic solution (Fluka, Buchs,

Switzerland), L-glutamine (2 mmol/L, Fluka), hydrocortisone (10 µg/ml, Fluka), and N6,2'-O-

dibutyryladenosine-3',5'-cyclic monophosphate sodium salt (25 µg/ml, Fluka) for up to 11

passages. Cells were grown in a humidified atmosphere at 37°C and 5% CO2.

Proliferation, migration, and tube formation assays were performed as described (Kajiya et al.

2005). Cells were incubated with 100 ng/ml human recombinant EGF (BD Biosciences), 20

ng/ml recombinant human VEGF-A165 (kindly provided by the National Cancer Institute,

Bethesda, MD), 1.2 µg/ml Pichia Pastoris-derived VEGF-C (kindly provided by Dr. K.

Ballmer-Hofer, Paul Scherrer Institute, Switzerland); 10 µM GW2974 (Sigma-Aldrich) or

equal amounts of DMSO as negative control. In additional experiments, cells were incubated

with 10 µg/ml of recombinant humanized monoclonal antibody 2C4 (Pertuzumab, kindly

provided by Roche Diagnostics GmbH, Mannheim, Germany) or 10 µg/ml of an EGFR

blocking antibody (Calbiochem, Darmstadt, Germany). Mouse IgG (Santa Cruz

Biotechnology, Heidelberg, Germany) was used as control.

Page 84: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

83

For proliferation assays, LECs (1.25 to 1.5 x 103) were seeded into fibronectin (Chemicon

International, Temecula, CA) coated 96-well plates. Cells were treated with the different

compounds. After 72 hours, cells were incubated with 5-methylumbelliferylheptanoate

(MUH, Sigma-Aldrich) as described (Detmar et al. 1992). The intensity of fluorescence,

proportional to the number of viable cells, was measured using a SpectraMax Gemini EM

microplate reader (Bucher Biotec AG, Basel, Switzerland). Ten wells per condition were

analyzed. For migration assays, two parallel lines and one orthogonal line were scratched in

confluent LEC cultures, to remove LECs without damaging the fibronectin coating. Pictures

of the two crosses per well were taken immediately after scratching (T0) and after 16 hours of

compound incubation (T16) with a Zeiss AxioCam-MRm (Zeiss). The quantitative analysis of

the non-closed area was performed using Photoshop C3. Three wells per condition were

analyzed.

For tube formation assays, confluent LEC monolayers were overlaid with collagen type I gels

(1 mg/ml; Cohesion, Palo Alto, CA) as described (Kajiya et al. 2005), containing the studied

compounds or their solvent only. Tube-like structure formation was evaluated for up to 20

hours. Three pictures of hot-spots were taken per well, and tube length was analyzed using the

IP Lab software as described (Kajiya et al. 2005). Three wells per condition were used. For all

in vitro studies, three independent experiments were performed. Statistical analyses were

performed using the two-tailed unpaired Student’s t-test (Graph Pad Prism 4).

4.2.3.4 Immunostaining of cultured human LECs

Dermal lymphatic endothelial cells were fixed in 4% paraformaldehyde (PFA) at 4°C, washed

in PBS, stained with rabbit anti-human EGFR antibody (Abcam, Cambridge, UK) and and a

secondary antibody conjugated with Alexa 594. Cell nuclei were counterstained with

Page 85: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

84

bisbenzimide. Specimens were examined with a Zeiss Axioskop 2 mot plus. All images were

captured with a Zeiss AxioCam-MRm.

4.2.3.5 Immunoblotting

For Western blot analyses of EGFR protein expression, confluent cultures of LEC were

homogenized in lysis buffer (Kajiya et al. 2005). The protein concentrations were determined

using the NanoOrange Protein Quantification Kit (Molecular Probes, Eugene, OR). The

lysates (100 µg of total protein) were then subjected to SDS-polyacrylamide gel

electrophoresis using NuPAGE 10% BT gels, 1.0 mm, 12 well, and NuPAGE MES SDS

running buffer (20×; Invitrogen). The proteins were transferred onto Trans-Blot Transfer

Medium pure nitrocellulose membranes (BioRad, Hercules, CA) for immunoblot analysis.

Blocking was performed with 5% non-fat dry milk in 0.1% Tween20 (Sigma) in PBS,

followed by immunoblotting with a rabbit anti-human EGFR antibody (Abcam). Specific

binding was detected by the ECL Plus Western Blotting Detection System (GE Healthcare,

Buckinghamshire, UK). Equal loading was confirmed using an antibody against β-actin

(Sigma).

4.2.3.6 In vivo matrigel lymphangiogenesis assay

FVB wild-type mice (female, 9-weeks old, n=5 per group; Charles River, Sulzbach, Germany)

were subcutaneuosly injected with 500 µl of growth factor reduced Matrigel (Becton

Dickinson), containing 1 µg/ml of VEGF-C or 1 µg/ml of EGF (BD Biosciences), or an equal

volume of PBS. After 15 days, tissues were excised, embedded and frozen in optimal cutting

temperature (OCT) compound (Sakura Finetek, Zoeterwoude, Netherlands).

Immunofluorescent staining for mouse LYVE-1 (Angiobio, Del Mar, CA) and CD31 (BD

Pharmingen) was performed using corresponding secondary antibodies labeled with Alexa-

Page 86: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

85

Fluor 488 or 594 (Molecular Probes). Cell nuclei were counterstained with Hoechst

bisbenzimide. The stained sections were examined with a Zeiss Axioskop 2 mot plus. All

images were captured with a Zeiss AxioCam-MRm. Vessel area, density and size were

evaluated with Photoshop C3. For statistical analyses, the two-tailed unpaired student’s t-test

was used (Graph Pad Prism 4). The animal study was approved by the Veterinaeramt des

Kantons Zuerich, Zuerich, Switzerland, permission number 123/2005.

4.2.3.7 Immunohistochemistry

Tail and ear skin paraffin sections from 7-days old keratin14-amphiregulin transgenic mice

(K14-ARtg) (Cook et al. 1997) and from wildtype FVB mice were incubated at 60°C for four

hours, dewaxed in xylene, hydrated and processed for immunohistochemistry for LYVE-1

(goat biotinylated anti-mouse LYVE-1 antibody; R&D Systems). The stained sections were

examined with a Zeiss Axioskop 2 mot plus. All images were captured with a Zeiss AxioCam-

MRm. Vessel area, density and size were evaluated with Photoshop C3. For statistical

analyses, the two-tailed unpaired Student’s t-test was used (Graph Pad Prism 4). Human skin

sections were fixed in acetone for 10 minutes. Double immunofluorescent staining for

podoplanin (D2-40, Signet, Dedham, MA) and EGFR (Abcam) was performed using

corresponding secondary antibodies labeled with Alexa-Fluor 488 or 594 (Molecular Probes).

Cell nuclei were counterstained with Hoechst bisbenzimide or hematoxylin.

Page 87: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

86

4.2.4 Results

4.2.4.1 Blockade of the EGF-EGFR/ErbB2 pathway inhibits the formation of

CD31+/LYVE-1+ vessel-like structures in embryoid bodies

We used our previously established mouse embryoid body (EB) vascular differentiation assay

to investigate the effects of several candidate small molecules on the development of

CD31+/LYVE-1+, lymphatic vessel-like structures. Treatment was started at day 14 after

initiation of the EBs, and was performed for four days. Compounds were added alone or in

combination with all-trans-retinoic acid (RA), cAMP and VEGF-C, which we have recently

found to potently promote lymphatic differentiation in EBs (Marino et al. manuscript

submitted).

EBs incubated with the mixture of RA, cAMP and VEGF-C showed an increased area and an

increased number of CD31+/LYVE-1+ vessel-like structures as compared to control EBs

(Figure 4.2.1 A-B). Incubation with SU5416, a VEGFR-2 tyrosine kinase inhibitor, with the

calcium channel inhibitors felodipine and nicardipine, or with the EGFR/ErbB2 tyrosine

kinase inhibitors genistein and GW2974, significantly reduced the CD31+/LYVE-1+ area of

the EBs. These effects were seen after incubation with the inhibitors alone and after

incubation with the inhibitors together with the mixture of RA, cAMP and VEGF-C (Figure

4.2.1 A). All of the five compounds tested also reduced the number of CD31+/LYVE-1+

vessel-like structures (Figure 4.2.1 B) when added to the EBs alone. In combination with RA,

cAMP and VEGF-C, only genistein, GW2974 and SU5416 showed significant inhibitory

activity (Figure 4.2.1 B). All compounds, alone or in combination with RA/cAMP/VEGF-C,

also slightly reduced the CD31+/LYVE-1- area and structure number (data not shown).

Page 88: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

87

Figure 4.2.1 The EGFR/ErbB2 pathway is involved in the formation of CD31+/LYVE-1+ vessel like

structures in EBs. Small molecules (10 µM) were added to the EBs, starting at day 14 after initiation, for four

days in combination or not with a mixture of all-trans-retinoic acid (RA), cAMP and VEGF-C. EBs exposed to

RA+cAMP+VEGF-C showed an increase of the CD31+/LYVE-1+ area and the number of vessel-like structures

as compared to control EBs (A and B). SU5416, felodipine, nicardipine, genistein and GW2974, alone or in

combination with RA+cAMP+VEGF-C, significantly reduced the LYVE-1+/CD31+ area (A). All 5 small

molecules also inhibited vessel-like structure formation as compared to medium only (B). In contrast, only

genistein, GW2974 and SU5416 significantly inhibited vessel like structure formation when used in combination

with RA+cAMP+VEGF-C (B). Data are expressed as mean values + SEM (n=9), *p<0.05; **p<0.01;***

p<0.001.

Whereas incubation of EBs with the mixture of RA, cAMP and VEGF-C increased the

formation of CD31+/LYVE-1+ vessel-like structures (Figure 4.2.2 B-B’’), compared with

control EBs (Figure 4.2.2 A-A’’), addition of GW2974 to the RA/cAMP/VEGF-C mixture

almost completely prevented the formation of CD31+/LYVE-1+ vessel-like structures, and

only clusters of CD31+/LYVE-1+ single cell were detected (Figure 4.2.2 C-D’’).

Page 89: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

88

Because inhibition of the EGFR/ErbB2 tyrosine kinas reduced the formation of

CD31+/LYVE-1+ vessel-like structures in EBs we next investigated whether epidermal

growth factor (EGF), an activating legend of the EGFR/ErbB2, might promote vessel

formation. EBs exposed to EGF for 4 days showed a significant increase in the EB area

occupied by CD31+/LYVE-1+ structures (Figure 4.2.3 A) and in the number of these

structures (Figure 4.2.3 B), as compared with controls. VEGF-C, used as a positive control,

showed a similar effect. The EGF effect was completely prevented by addition of GW2974

(Figure 4.2.3 A, B). Microscopical imaging revealed that both VEGF-C and EGF promoted

the formation of CD31+/LYVE-1+ vessel-like structures (Figure 4.2.3 D-E'') as compared to

controls (Figure 4.2.3 C-C’’). Addition of GW2974 together with EGF prevented the

formation of CD31+/LYVE-1+ vessel-like structures and only single CD31+/LYVE-1+ cells

clusters were found (Figure 4.2.3 F-F’’). Taken together, these data indicate a possible role of

EGF via EGFR/ErbB2 signalling in the formation of lymphatic vessel-like structures.

Page 90: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

89

Figure 4.2.2 GW2974 inhibits the formation of CD31+/LYVE-1+ vessel-like structures in EBs. Double

immunofluorescent staining for CD31 (red) and LYVE-1 (green) showed the formation of CD31+/LYVE-1+

vessel-like structures in control EBs (medium, A-A’’). Four days of EB incubation with RA+cAMP+VEGF-C

increased the formation and the complexity of structure networks (B-B’’) as compared with control. Co-

treatment of EBs with RA+cAMP+VEGF-C and GW2974 strongly inhibited the formation of those structures;

only single CD31+/LYVE-1+ cell clusters are present in the EBs (C-D’’). Scale Bars: 100 μM.

Page 91: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

90

Figure 4.2.3 EGF promotes the formation of CD31+/LYVE-1+ vessel-like structures in EBs. VEGF-C,

EGF, GW2974 or DMSO were added or not to EBs, starting at day 14 after initiation, for four days. Exposure to

EGF, as well as to VEGF-C, strongly increased the CD31+/LYVE-1+ area (A) and the number of vessel-like

structures (B) as compared with control. The EGF effect was inhibited by GW2974 but not by DMSO (A, B).

Double immunofluorescent staining for CD31 (red) and LYVE-1 (green) showed that EGF (E-E’’), together with

VEGF-C (D-D’’) promoted CD31+/LYVE-1+ vessel-like structure formation as compared to controls (C-C’’).

Exposure to a combination of EGF and GW2974 inhibited CD31+/LYVE-1+ vessel formation; only

CD31+/LYVE-1+ single cells clusters are visible (F-F’’). Data are expressed as mean values (n=9) + SEM;

*p<0.05; **p<0.01;*** p<0.001Scale bar: 50 µm.

Page 92: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

91

4.2.4.2 EGFR is expressed by human LECs in vitro and in vivo

The expression of EGFR on human blood vascular endothelial cells has been previously

reported (Sini et al. 2005). In contrast, EGFR expression on human lymphatic endothelial cells

has not been studied. Immunofluorescence and immunoblot analyses of human LECs in vitro

revealed that LECs express EGFR, although at rather low levels (Figure 4.2.4 A-C). The

expression of EGFR was slightly induced following 24h of EGF exposure (Figure 4.2.4 A-C).

Double immunofluorescent staining of human skin for EGFR and podoplanin revealed that

podoplanin-positive dermal lymphatic vessels express low levels of EGFR in vivo (Figure

4.2.4 D-F; arrowhead). Low levels of EGFR expression were also detected in podoplanin-

negative blood vessels (Figure 4.2.4 D-F; arrow).

Figure 4.2.4 Human LECs express EGFR in vitro and in vivo. Immunofluorescent analysis (A-B) and

immunoblot (C) for EGFR showed that human LECs express low levels of EGFR in vitro. The expression of the

receptor was slightly induced following 24h of EGF exposure (A-C). Double immunofluorescent analysis of

human skin for EGFR (red) and podoplanin (green) revealed that dermal lymphatic vessels express low levels of

EGFR (D-F arrow heads). Podoplanin negative blood vessels also express low levels of EGFR (D-F arrows).

Scale bars 50 μm.

Page 93: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

92

4.2.4.3 EGF-EGFR/ErbB2 signalling promotes in vitro lymphangiogenesis

Lymphangiogenesis is a complex process that involves proliferation, migration and vessel

formation of LECs. To further characterize the potential role of EGFR/ErbB2 signalling in

these processes, we next treated human LECs with GW2974 and performed in vitro

proliferation, migration and tube formation assays. Migration was evaluated in an in vitro

"scratch" wound healing assay. We found that GW2974 did not inhibit LEC proliferation nor

migration (Figure 4.2.5 A, B), but that it potently inhibited tube formation in vitro (Figure

4.2.5 C), as compared to untreated controls. VEGF-A or VEGF-C, used as positive controls,

showed a significant activation of these processes (Figure 4.2.5 A-C).

Figure 4.2.5 GW2974 inhibits human lymphatic endothelial cell tube formation in vitro. Human dermal

LECs (hLECs) were used to study cell proliferation (A), migration (B) and tube formation (C). Incubation with

VEGF-A significantly promoted hLEC proliferation and migration (A, B). Incubation with GW2974 did not

inhibit hLECs proliferation (A) or migration (B). Incubation with VEGF-C significantly promoted hLEC tube

formation (C). In contrast, GW2974 significantly inhibited tube formation as compared with control (C). Data are

expressed as mean values (n=3) + SEM; t-test: *p<0.05; *** p<0.001. AFU: absolute fluorescent units.

Page 94: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

93

We next studied the in vitro effects of EGF on LECs. Incubation with EGF had no major

effect on LEC proliferation (Figure 4.2.6 A) but significantly stimulated migration and tube

formation (Figure 4.2.6 B-C). Because small molecule tyrosine kinase inhibitors may inhibit a

number of tyrosine kinases, in addition to their most specific target, we next used a specific

EGFR blocking antibody (EGFRbAB) and an ErbB2 heterodimeryzation blocking antibody

(pertuzumab). The stimulating effects of EGF on cell migration were significantly inhibited by

EGFRbAB and by pertuzumab, and even more potently by a combination of both antibodies

(Figure 4.2.6 B). GW2974 also inhibited the EGF effect (Figure 4.2.6 B). No major effect on

LEC migration was observed after incubation with either the blocking antibodies alone, with

GW2974 or with the IgG controls (Figure 4.2.6 B). Similarly, the stimulating effect of EGF

on LEC tube formation was inhibited by EGFRbAB and by pertuzumab (Figure 4.2.6 C).

GW2974 also inhibited tube formation alone or in combination with EGF (Figure 4.2.6 C).

The blocking antibodies alone or the IgG controls had no major effects on tube formation

(Figure 4.2.6 C).

Page 95: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

94

Figure 4.2.6 EGF, via EGFR/ErbB2,

promotes human lymphatic endothelial cell

migration and tube formation in vitro.

Human dermal lymphatic endothelial cells

(hLECs) were used to study cell proliferation

(A), migration (B) and tube formation (C).

Incubation with VEGF-A or VEGF-C promoted

hLEC proliferation/migration (A, B) and tube

formation (C), respectively, as compared with

controls. EGF did not induce cell proliferation

(A) but it significantly induced hLEC migration

and tube formation as compared with controls

(B, C). The EGF effect was inhibited by

GW2974, by an EGFR blocking antibody

(EGFRbAB), by pertuzumab and by the

combination of the two antibodies (B, C).

GW2974 alone significantly inhibited tube

formation (C). The blocking antibodies alone as

well as the IgG control did not affect hLEC

migration (B) or tube formation (C). Data are

expressed as mean values (n=3) + SEM;

*p<0.05; **p<0.01;*** p<0.001. AFU:

absolute fluorescent units.

Page 96: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

95

4.2.4.4 EGF promotes lymphangiogenesis in vivo

We next investigated whether EGF might also have an effect on lymphatic vessel formation in

vivo. To this end, matrigel plugs containing EGF, VEGF-C or vehicle alone were injected

subcutaneously into the left inguinal area of wild type FVB mice. After 2 weeks, the matrigel

plugs and the associated skin were dissected and analyzed. We found a significantly increased

lymphatic vessel area (p=0.014), increased lymphatic vessel size (p=0.53) and increased

lymphatic vessel density (p=0.073) in the skin surrounding matrigel plugs that contained EGF

(Figure 4.2.7 A-C), as compared to those that contained PBS. The effects of EGF were largely

comparable to those of VEGF-C (Figure 4.2.7 A-C).

Figure 4.2.7 EGF promotes lymphangiogenesis in vivo. A matrigel plug containing PBS, VEGF-C or EGF

was injected subcutaneously into the left inguinal area of wild type mice. After 2 weeks, the plug and the

associated skin were dissected and quantitatively analyzed. VEGF-C significantly increased lymphatic vessel

area (A) and size (B) as compared with control PBS; the lymphatic density was not significantly increased (C).

EGF treatment significantly induced lymphatic vessels area (A) and it slightly increased size (B) and density (C).

Data are expressed as absolute values per matrigel plug; bar represents the median value.

Page 97: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

96

4.2.4.5 Transgenic overexpression of amphiregulin in mouse skin promotes

lymphangiogenesis in vivo

Amphiregulin is another specific ligand for EGFR/ErbB2. Recently, it has been shown that

transgenic mice that express amphiregulin under control of the K14 promoter in the skin,

develop a chronic, psoriasis-like skin inflammation early during life (Cook et al. 1997). We

next investigated whether chronic transgenic overexpression of amphiregulin in the mouse

skin might also promote lymphangiogenesis. We found an increased lymphatic vessel area

(Figure 4.2.8 A), size (Figure 4.2.8 B) and density (Figure 4.2.8 C) in both the ear skin and the

tail skin of seven-days old transgenic mice, as compared with wild type mice of the same age.

Immunohistochemical analysis of LYVE-1 expression in the tail skin (Figure 4.2.8 D, E) and

ear skin (Figure 4.2.8 F, G) revealed increased numbers of enlarged dermal lymphatic vessels

in K14/amphiregulin transgenic mice (Figure 4.2.8 E, G) as compared with wild type mice

(Figure 4.2.8 D, F).

Page 98: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

97

Figure 4.2.8 Transgenic overexpression of amphiregulin in mouse skin promotes lymphangiogenesis.

Seven-day old K14-amphiregulin (AR) transgenic mice presented an increased lymphatic vessel area (A), size

(B) and density (C) in both ear skin and tail skin, as compared with wild type (WT) mice (the increase is not

significant for lymphatic size in the tail sample, B). Immunohistochemical analysis for LYVE-1 in the tail (D, E)

and ear (F, G) skin showed enlarged and more dense dermal lymphatics in the transgenic mouse samples (E, G)

as compared with wild type animals (D, F arrow heads. Data are expressed as absolute values per animal, bar

represents the mean value. Scale bars 50 μm.

Page 99: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

98

4.2.5 Discussion

In a search for novel pathways involved in lymphatic vessel growth, we have investigated a

number of small molecular organic compounds for their effects on vascular development in

mouse embryonic stem cell-derived embryoid bodies (EBs). The results indicated that

epidermal growth factor receptor (EGFR/ErbB2) signalling might regulate lymphatic vessel

formation and conversely that EGF, via EGFR/ErbB2, promotes vessel growth. We also found

that human dermal LECs express low levels of EGFR both in vitro and in vivo and that

signalling of EGF via EGFR/ErbB2 promotes human LEC migration and tube formation.

Importantly, two independent in vivo studies reveal that EGFR signalling also promote

lymphangiogenesis in vivo.

Recently, a forward chemical genetics approach, using a chemical library containing 1,280

small organic molecules with well-defined pathway specificities, was applied to Xenopus

laevis tadpoles (Kalin et al. 2009). A phenotypic screen revealed compounds affecting either

blood vessel formation, lymphatic vessel formation or both. Among the compounds with

effects on vascular development, SU5416 (targeting the tyrosine kinase associated with

VEGFR-2) potently inhibited blood vessel formation, whereas GW2974, an EGFR/ErbB2

tyrosine kinase inhibitor, and the calcium channel blockers felodipine and nicardipine,

predominantly inhibited lymphatic vessel development. Genistein, another EGFR/ErbB2

tyrosine kinase inhibitor, affected both blood and lymphatic vessel formation. The lymphatic

phenotypes were associated with impaired lymph sac and lymph heart formation, and with

hypoplasia of the posterior lymphatic vessels (Kalin et al. 2009). These results indicated a

role of EGFR signalling in developmental lymphangiogenesis. However, the small organic

tyrosine kinase inhibitors also target other tyrosine kinases than EGFR, and the potential role

Page 100: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

99

of EGF/EGFR signalling for the growth and function of lymphatic vessels after

embryogenesis has remained unknown.

Mouse embryonic stem cell-derived EBs have been used to study (anti-)angiogenic properties

of small molecules and drugs (Wartenberg et al. 1998). Recently, we found that also

lymphatic vessel-like structures develop in EBs and that incubation of EBs with the

lymphangiogenic growth factor VEGF-C promotes formation of CD31+/LYVE-1+ lymphatic

vessel like structures (Liersch et al. 2006), indicating that EBs might be useful tools for

identifying compounds potentially modulating lymphangiogenesis. We tested the selected

compounds alone or in combination with a mixture of retinoic acid (RA), cAMP and VEGF-

C, since we recently found that RA in combination with cAMP potently promote the

development of lymphatic vessel-like structures in EBs (Marino et al., submitted). Although

the calcium channel blocking compounds felodipine and nicardipine, as well as genistein, also

reduced the number of CD31+/LYVE-1+ vessel-like structures in EBs, incubation with

GW2974 showed the strongest inhibitory effect in the EBs. In fact, GW2974 almost

completely prevented the formation of lymphatic vessel like structures, and only single

CD31+/LYVE-1+ cells were observed, even when EBs were incubated in the

RA/cAMP/VEGF-C mixture.

EGFR family proteins (Riese and Stern 1998) play important roles in the regulation of cell

growth, proliferation, and differentiation of various cell types. In particular, mice lacking the

EGFR (ErbB1) die before implantation or at mid-late gestation (depending on the genetic

background), due to placental malformations and severe defects of various epithelial cells

(Wong 2003). In particular, it has been shown that EGFR may regulate angiogenesis both

directly (Baker et al. 2002) and indirectly (Kumar and Yarmand-Bagheri 2001). Moreover,

Page 101: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

100

EGFR ligands such as EGF and TGFα are potent pro-angiogenic factors (Schreiber et al.

1986), in part via upregulation of VEGF-A expression in epithelial cells (Detmar et al. 1994)

The EGFR has been reported to be expressed by quiescent and by tumor-associated blood

vascular endothelial cells (Sini et al. 2005; Amin et al. 2006). Importantly, we detected EGFR

expression in human dermal lymphatic endothelial cells both in vitro and in vivo. These

findings indicate that the effects of EGF (and of amphiregulin) on lymphatic vessel activation

are, at least in part, mediated directly via EGFR activation. Recently, it has been reported that

proliferation of cultured mouse mesenteric LEC was induced by EGF (via Akt and ERK1/2)

and inhibited by AEE788, a small molecule that targets EGFR and VEGFR2 TKs (Rebhun et

al. 2006). EGF has also been reported to stimulate proliferation of bovine LECs (Liu and He

1997). In contrast, we did not observe any significant effect of EGF treatment, or of

incubation with the EGFR inhibitor GW2974, on the in vitro proliferation of human dermal

lymphatic endothelial cells in the present study. These differences might be due to the

different culture conditions, to different tissue origin of the LECs, or to species-specific

differences in the response of lymphatic endothelium to growth stimulation. Importantly,

however, our in vitro studies revealed that activation of the EGFR potently promotes LEC

migration and tube formation, and that - conversely - blockade of EGFR signalling via direct

antibody inhibition and via tyrosine kinase inhibition reduce migration and tube formation.

Our study is the first to reveal that activation of the EGFR promotes lymphangiogenesis in

vivo. We have used two independent experimental mouse models to investigate the role of this

receptor in lymphatic vessel formation. Implantation of an EGF containing matrigel plug

resulted in an increased number of enlarged lymphatic vessels in the surrounding skin after 14

days. The matrigel plug assay is a widely used and established (lymph)-angiogenic assay.

However, implantation also leads to an inflammatory response of the host tissue, with possible

Page 102: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

101

confounding effects by the inflammatory cells. Thus, we investigated a second mouse model

with chronic overexpression of another EGFR ligand, amphiregulin, by transgenic

keratinocytes within the epidermis. We initially studied older K14/amphiregulin mice at an

age of 3 weeks, and found dramatic enlargement of the cutaneous lymphatic vasculature (data

not shown). However, at this time point, the mice had developed a strong, psoriasis-like skin

inflammation, in agreement with the published phenotype (Cook et al. 1997). Because

psoriasis skin lesions in humans, and psoriasis-like skin lesions in mice are associated with

massive lymphatic hyperplasia (Kunstfeld et al. 2004), we then studied the vasculature of

young, 1-week old K14-amphiregulin transgenic mice (at a time point when there was no

massive inflammation detectable). These investigations revealed that overexpression of

amphiregulin promoted dermal lymphangiogenesis. Taken together, the results indicate a role

of EGFR signalling in lymphatic vessel growth in vivo.

Is the EGF effect on lymphatic endothelial cells mediated directly or indirectly? We were

unable to detect EGFR expression neither on mRNA level in ex vivo isolated mouse colon

lymphatic endothelial cells nor on protein level in mouse dermal lymphatic vessels (data not

shown); thus, the pro-lymphangiogenic effect of EGF and amphiregulin in vivo might be - at

least partially - due to an indirect effect mediated by other cells types that might also be

present in the EB assay. In fact, it has been shown that EGFR/ErbB2 signalling leads to the

expression of angiogenic growth factor, such as VEGF-A in keratinocytes and cancer cells

(Detmar et al. 1994; Dvorak et al. 1995; Kumar and Yarmand-Bagheri 2001; Al-Nedawi et al.

2009). In contrast, the effect of EGF on the migration and tube formation of human dermal

LECs was most likely mediated directly since these cells express the EGFR and since no other

cell type was present in these cultures. Moreover, the levels of VEGF-A and VEGF-C were

below detection limit in LEC cultures treated with EGF, as evaluated by ELISA (data not

shown).

Page 103: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

102

EGFR and ErBb2 are frequently upregulated in human carcinomas and support proliferation

and survival of cancer cells (Yarden and Sliwkowski 2001). Small molecule inhibitors such as

erlotinib and blocking antibodies such as cetuximab, that target EGFR, are in clinical use for

some types of cancer such as lung cancers (Herbst and Sandler 2008). Inhibition of tumor

associated-angiogenesis by EGFR blockade has been reported (Petit et al. 1997; Perrotte et al.

1999), and combination therapies blocking EGF and VEGF signalling are currently under

investigation as a potential strategy to overcome resistance to anti-angiogenic therapy.

Importantly, patients who undergo EGFR blocking treatment often develop an acute skin rush

and skin inflammation (Tsimboukis et al. 2009), and development of peripheral edema has

been reported (Perez-Soler and Saltz 2005; Von Minckwitz et al. 2005). Based upon our

results, analysis of patient biopsies would be of great interest to identify possible effect of

EGFR inhibition on tumor-associated lymphangiogenesis.

In conclusion, the present study indicates that small inhibitory molecules (e.g. erlotinib) or

blocking antibodies (e.g. cetuximab, pertuzumab) targeting EGFR/ErbB2 signalling might be

potential candidates for the treatment of pathological lymphangiogenesis in cancer and other

diseases.

Page 104: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

103

5 CONCLUSIONS AND OUTLOOK

The present work has aimed at identifying molecular regulators of the earliest steps of lymphatic

vascular development. So far, this task has proven challenging using classical in vivo genetic

manipulation techniques due to the fact that several candidate gene mutations have resulted in

mouse embryo lethality. The use of mouse embryonic stem cell-derived embryoid bodies, an in

vitro model that largely mimics mouse embryogenesis, has allowed us to circumvent this

problem.

Using the embryoid body vascular differentiation assay, we have identified retinoic acid (RA) as

a novel mediator of lymphatic competence and commitment. RA alone, and in combination with

cAMP, potently up-regulated LYVE-1 expression in CD31+ vascular structures. Indeed, these

LYVE-1+, lymphatically competent endothelial cells emerged from the pre-existing CD31+

blood vascular endothelium, supporting the centrifugal theory of lymphatic development

proposed by Sabin in 1902 (Sabin 1902). However, the combination of RA and cAMP also

induced the formation of rare CD31+/LYVE-1+/Prox1+ cell clusters that emerged

independently from the CD31+ vessel-like networks. Thus, also supporting the centripetal

theory proposed by Huntington in 1910 (Huntington and McClure 1910). Our studies also

demonstrated that retinoic acid receptor (RAR)-was expressed by the endothelial cell of the

embryonic cardinal veins in mouse embryos and that Ro 41-5253, a RAR- antagonist

inhibited the induction of lymphatic vascular structures by RA, suggesting that RA activity is

most likely mediated via RAR-α. The potentiation of RA’s effects by cAMP and the inhibition

by H89 (a cAMP-dependent PkA inhibitor) indicate that RA’s effects are mediated by a

pathway that also involves cAMP and PkA. Importantly, in mouse embryos increased levels of

RA in utero upregulated LYVE-1 expression in the endothelial cells of the cardinal veins and

Page 105: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

104

jugular lymph sacs, and increased the number of CD31+/Prox1+ lymphatic endothelial cells. In

contrast, in utero injection of Ro 41-5253 decreased expression of LYVE-1 and the number of

CD31+/Prox1+ lymphatic endothelial cells in the same vascular regions.

Moreover, exposure of Xenopus embryos to RA induced the expression of VEGFR-3 and

LYVE-1, thus conferring lymphatic competence to the developing vasculature.

Retinoids, such as 13-cis-RA, are widely used via both topical (Orfanos et al. 1997) and

systemic administration (DiGiovanna 2001) for the treatment of various conditions such as

acne, psoriasis, postinflammatory hyperpigmentation, photodamaged skin, leukemia, disorders

of keratinisation, premalignant skin lesions, cutaneous T-cell lymphoma, and dissecting

cellulitis of the scalp (Saurat 1998). Recently, retinoids are being considered as agents to treat

autoimmune diseases, such as multiple sclerosis (Pino-Lagos et al. 2008) and Alzheimer’s

disease (Shudo et al. 2009), due to the fact that they can, in general, exert immunosuppressive

effects. Importantly, therapeutic efficacy of retinoids has been reported for the treatment of

persistent solid facial edema, a complication of acne vulgaris (Thielitz and Gollnick 2008;

Manolache et al. 2009). In contrast, retinoic acid syndrome, a clinical syndrome that occurs after

treatment of leukemia (Leelasiri et al. 2005) with all-trans-retinoic acid, is very often associated

with edema formation and weight gain. Since lymphatic vessels are known to be involved in

inflammatory processes, as well as in edema formation and obesity (Cueni and Detmar 2008),

this clinical evidence suggests a possible link between RA treatment and lymphatic vessel

growth and function. Indeed, preliminary results in a pilot study on adult mice indicate that

intraperitoneal injection of RA induces dermal lymphangiogenesis (data not shown). Thus, it

will be of great interest to determine whether retinoids might modulate the growth of new

lymphatic vessels in pathological conditions like lymphedema. In this respect, mouse models of

e.g. tail lymphedema (Jin et al. 2008) might represent a suitable in vivo approach to evaluate the

effect of RA on the regression of the pathology.

Page 106: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

105

The second part of this thesis focused on using the embryoid body model to identify novel

(anti)-lymphangiogenic pathways. To this end, embryoid bodies were treated with small organic

molecules with well-defined pathway specificities, and the formation of lymphatic vessel-like

structures was evaluated. The screening identified the small molecule GW2974, which targets

the tyrosine kinase associated with epidermal growth factor receptor (EGFR/ErbB2), as an

inhibitor of lymphatic vessel formation. Conversely, embryoid bodies treated with EGF, an

activating ligand for EGFR/ErbB2, displayed increased number of lymphatic vessel networks as

compared to control embryoid bodies. We found that human dermal lymphatic endothelial cells

(hLECs) express low levels of EGFR. GW2974, as well as EGFR/ErbB2 blocking antibodies,

inhibited in vitro migration and tube formation of hLECs whereas EGF significantly induced

those processes. In vivo analysis of mice subcutaneously implanted with EGF containing

matrigel plugs, as well as of mice expressing amphiregulin (another EGFR/ErbB2 ligand) under

the control of K14 promoter, revealed that the activation of the EGFR/ErbB2 signalling pathway

induces dermal lymphangiogenesis. Taken together, our results suggest that targeting EGFR

might represent a novel strategy to interfere with lymphangiogenesis in pathological conditions.

Further in vivo studies, such as blocking EGFR in tumor bearing mice, could prove valuable in

defining the therapeutic potential of EGFR antagonists to inhibit tumor metastasis. Moreover,

since small molecules such as erlotinib and blocking antibodies such as cetuximab that target

EGFR are already in clinical use for some types of cancer such as lung cancers (Herbst and

Sandler 2008), the analysis of patient tumor biopsies would be of great interest to identify a

possible effect of EGFR inhibition on tumor-associated lymphangiogenesis.

In conclusion, mouse embryonic stem cell-derived embryoid bodies have allowed us to discover

novel processes regulating lymphatic differentiation and lymphangiogenesis. These findings

open up the way for future research projects with the aim of better understanding the molecular

mechanisms that control growth and function of the lymphatic vascular system.

Page 107: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

106

6 ACKNOWLEDGMENTS

I wish to thank Michael Detmar for giving me the opportunity of doing my PhD in such an

interesting field and at such an honourable institution as ETH. Thanks for patiently

accompanying me through the fabulous and exciting world of science.

Many thanks go to Prof. Cornelia Halin for being co-referee of my thesis and for her always

helpful suggestions.

I thank Prof. André Braendli and Dr. Vasilios Debouras for providing the Xenopus techniques,

helping in performing the experiments and for revising the manuscript: “A role for all-trans-

retinoic acid in the early steps of lymphatic vascular system development”.

I would like to thank my two students Yvonne Angehrn and Sabrina Riccardi for their

dedication to the “EB factories” and the great time together inside and outside the lab!

For the study: “Activation of the epidermal growth factor receptor promotes lymphangiogenesis

in vitro and in vivo” I thank Dr. Baenziger-Tobler, Yvonne Angehrn and Sabrina Riccardi for

their contribution, Prof. M. Pittelkow for providing the K14-AR mouse samples and Mr. M.

Hasmann (Roche Diagnostics GmbH) for providing the human ErbB2 antibody, pertuzumab.

Special thanks go to Deena and Steven, our precious native English speaking post docs that

“volunteered” to correct this thesis, fishing and hunting out Italian expressions and misplaced

commas!

Immense thanks go to the wonderful “Detmar&Halin lab”. All present and former members

have not just been my colleagues: they have been and are my “Swiss Family”, my irreplaceable

Page 108: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

107

friends. Exceptional thanks go to Giorgia, Jay, Christian, Matthias, Tanja, Max, Steven, Martin,

Benni and Masha. Those four years with you guys have been unforgettable. Moreover,

uncountable thanks go to one lab member above all: my office-mate and best friend Milica that

intensively shared with me Ph.D. happiness and frustration as well as very precious life

moments.

At last but not least I would like to thank my beloved fiancé Gianluca, my little sister Roberta

and my parents for always believing in me and supporting any of my (crazy or intelligent)

decisions and ideas. Without them I would not be who, what and where I am! I love you all so

much!

Page 109: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

108

7 REFERENCES

Abtahian, F., Guerriero, A., Sebzda, E., Lu, M.M., Zhou, R., Mocsai, A., Myers, E.E., Huang, B., Jackson, D.G., Ferrari, V.A. et al. 2003. Regulation of blood and lymphatic vascular separation by signaling proteins SLP-76 and Syk. Science 299: 247-251.

Achen, M.G., Jeltsch, M., Kukk, E., Makinen, T., Vitali, A., Wilks, A.F., Alitalo, K., and Stacker, S.A. 1998. Vascular endothelial growth factor D (VEGF-D) is a ligand for the tyrosine kinases VEGF receptor 2 (Flk1) and VEGF receptor 3 (Flt4). Proc. Natl. Acad. Sci. U. S. A. 95: 548-553.

Achen, M.G., McColl, B.K., and Stacker, S.A. 2005. Focus on lymphangiogenesis in tumor metastasis. Cancer Cell 7: 121-127.

Al-Nedawi, K., Meehan, B., Kerbel, R.S., Allison, A.C., and Rak, J. 2009. Endothelial expression of autocrine VEGF upon the uptake of tumor-derived microvesicles containing oncogenic EGFR. Proc. Natl. Acad. Sci. U. S. A. 106: 3794-3799.

Al-Rawi, M.A., Watkins, G., Mansel, R.E., and Jiang, W.G. 2005. The effects of interleukin-7 on the lymphangiogenic properties of human endothelial cells. Int. J. Oncol. 27: 721-730.

Alitalo, K., Tammela, T., and Petrova, T.V. 2005. Lymphangiogenesis in development and human disease. Nature 438: 946-953.

Amin, D.N., Hida, K., Bielenberg, D.R., and Klagsbrun, M. 2006. Tumor endothelial cells express epidermal growth factor receptor (EGFR) but not ErbB3 and are responsive to EGF and to EGFR kinase inhibitors. Cancer Res. 66: 2173-2180.

Angeli, V., Ginhoux, F., Llodra, J., Quemeneur, L., Frenette, P.S., Skobe, M., Jessberger, R., Merad, M., and Randolph, G.J. 2006. B cell-driven lymphangiogenesis in inflamed lymph nodes enhances dendritic cell mobilization. Immunity 24: 203-215.

Apfel, C., Bauer, F., Crettaz, M., Forni, L., Kamber, M., Kaufmann, F., LeMotte, P., Pirson, W., and Klaus, M. 1992. A retinoic acid receptor alpha antagonist selectively counteracts retinoic acid effects. Proc. Natl. Acad. Sci. U. S. A. 89: 7129-7133.

Asellius, G. 1627. De lactibus sive lacteis venis. Milan, Mediolani. Aslakson, C.J. and Miller, F.R. 1992. Selective events in the metastatic process defined by analysis

of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 52: 1399-1405.

Ayadi, A., Zheng, H., Sobieszczuk, P., Buchwalter, G., Moerman, P., Alitalo, K., and Wasylyk, B. 2001. Net-targeted mutant mice develop a vascular phenotype and up-regulate egr-1. EMBO J. 20: 5139-5152.

Backhed, F., Crawford, P.A., O'Donnell, D., and Gordon, J.I. 2007. Postnatal lymphatic partitioning from the blood vasculature in the small intestine requires fasting-induced adipose factor. Proc. Natl. Acad. Sci. U. S. A. 104: 606-611.

Bain, G., Kitchens, D., Yao, M., Huettner, J.E., and Gottlieb, D.I. 1995. Embryonic stem cells express neuronal properties in vitro. Dev. Biol. 168: 342-357.

Bajenoff, M., Granjeaud, S., and Guerder, S. 2003. The strategy of T cell antigen-presenting cell encounter in antigen-draining lymph nodes revealed by imaging of initial T cell activation. J. Exp. Med. 198: 715-724.

Baker, C.H., Kedar, D., McCarty, M.F., Tsan, R., Weber, K.L., Bucana, C.D., and Fidler, I.J. 2002. Blockade of epidermal growth factor receptor signaling on tumor cells and tumor-associated endothelial cells for therapy of human carcinomas. Am. J. Pathol. 161: 929-938.

Baldwin, M.E., Halford, M.M., Roufail, S., Williams, R.A., Hibbs, M.L., Grail, D., Kubo, H., Stacker, S.A., and Achen, M.G. 2005. Vascular endothelial growth factor D is dispensable for development of the lymphatic system. Mol. Cell. Biol. 25: 2441-2449.

Page 110: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

109

Baluk, P., Fuxe, J., Hashizume, H., Romano, T., Lashnits, E., Butz, S., Vestweber, D., Corada, M., Molendini, C., Dejana, E. et al. 2007. Functionally specialized junctions between endothelial cells of lymphatic vessels. J. Exp. Med. 204: 2349-2362.

Baluk, P., Tammela, T., Ator, E., Lyubynska, N., Achen, M.G., Hicklin, D.J., Jeltsch, M., Petrova, T.V., Pytowski, B., Stacker, S.A. et al. 2005. Pathogenesis of persistent lymphatic vessel hyperplasia in chronic airway inflammation. J. Clin. Invest. 115: 247-257.

Banerji, S., Ni, J., Wang, S.X., Clasper, S., Su, J., Tammi, R., Jones, M., and Jackson, D.G. 1999. LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific receptor for hyaluronan. J.Cell Biol. 144: 789-801.

Banziger-Tobler, N.E., Halin, C., Kajiya, K., and Detmar, M. 2008. Growth hormone promotes lymphangiogenesis. Am. J. Pathol. 173: 586-597.

Barsky, S.H., Baker, A., Siegal, G.P., Togo, S., and Liotta, L.A. 1983. Use of anti-basement membrane antibodies to distinguish blood vessel capillaries from lymphatic capillaries. Am. J. Surg. Pathol. 7: 667-677.

Beasley, N.J., Prevo, R., Banerji, S., Leek, R.D., Moore, J., van Trappen, P., Cox, G., Harris, A.L., and Jackson, D.G. 2002. Intratumoral lymphangiogenesis and lymph node metastasis in head and neck cancer. Cancer Res. 62: 1315-1320.

Bekker, M.N., van den Akker, N.M., Bartelings, M.M., Arkesteijn, J.B., Fischer, S.G., Polman, J.A., Haak, M.C., Webb, S., Poelmann, R.E., van Vugt, J.M. et al. 2006. Nuchal edema and venous-lymphatic phenotype disturbance in human fetuses and mouse embryos with aneuploidy. J. Soc. Gynecol. Investig. 13: 209-216.

Bermingham-McDonogh, O., Oesterle, E.C., Stone, J.S., Hume, C.R., Huynh, H.M., and Hayashi, T. 2006. Expression of Prox1 during mouse cochlear development. J. Comp. Neurol. 496: 172-186.

Bjorndahl, M.A., Cao, R., Burton, J.B., Brakenhielm, E., Religa, P., Galter, D., Wu, L., and Cao, Y. 2005. Vascular endothelial growth factor-a promotes peritumoral lymphangiogenesis and lymphatic metastasis. Cancer Res. 65: 9261-9268.

Breiteneder-Geleff, S., Soleiman, A., Horvat, R., Amann, G., Kowalski, H., and Kerjaschki, D. 1999. [Podoplanin--a specific marker for lymphatic endothelium expressed in angiosarcoma]. Verh. Dtsch. Ges. Pathol. 83: 270-275.

Brice, G., Child, A.H., Evans, A., Bell, R., Mansour, S., Burnand, K., Sarfarazi, M., Jeffery, S., and Mortimer, P. 2005. Milroy disease and the VEGFR-3 mutation phenotype. J. Med. Genet. 42: 98-102.

Bromley, S.K., Thomas, S.Y., and Luster, A.D. 2005. Chemokine receptor CCR7 guides T cell exit from peripheral tissues and entry into afferent lymphatics. Nat. Immunol. 6: 895-901.

Burke, Z. and Oliver, G. 2002. Prox1 is an early specific marker for the developing liver and pancreas in the mammalian foregut endoderm. Mech. Dev. 118: 147-155.

Buttler, K., Ezaki, T., and Wilting, J. 2008. Proliferating mesodermal cells in murine embryos exhibiting macrophage and lymphendothelial characteristics. BMC. Dev. Biol. 8: 43.

Buttler, K., Kreysing, A., von Kaisenberg, C.S., Schweigerer, L., Gale, N., Papoutsi, M., and Wilting, J. 2006. Mesenchymal cells with leukocyte and lymphendothelial characteristics in murine embryos. Dev. Dyn. 235: 1554-1562.

Canon, E., Cosgaya, J.M., Scsucova, S., and Aranda, A. 2004. Rapid effects of retinoic acid on CREB and ERK phosphorylation in neuronal cells. Mol. Biol. Cell 15: 5583-5592.

Cao, R., Bjorndahl, M.A., Religa, P., Clasper, S., Garvin, S., Galter, D., Meister, B., Ikomi, F., Tritsaris, K., Dissing, S. et al. 2004. PDGF-BB induces intratumoral lymphangiogenesis and promotes lymphatic metastasis. Cancer Cell 6: 333-345.

Cao, Y. 2005a. Direct role of PDGF-BB in lymphangiogenesis and lymphatic metastasis. Cell Cycle 4: 228-230.

Cao, Y. 2005b. Opinion: emerging mechanisms of tumour lymphangiogenesis and lymphatic metastasis. Nat. Rev. Cancer 5: 735-743.

Page 111: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

110

Casley-Smith, J.R. 1980. The fine structure and functioning of tissue channels and lymphatics. Lymphology 13: 177-183.

Caunt, M., Mak, J., Liang, W.C., Stawicki, S., Pan, Q., Tong, R.K., Kowalski, J., Ho, C., Reslan, H.B., Ross, J. et al. 2008. Blocking neuropilin-2 function inhibits tumor cell metastasis. Cancer Cell 13: 331-342.

Chan, B.W., Chan, K.S., Koide, T., Yeung, S.M., Leung, M.B., Copp, A.J., Loeken, M.R., Shiroishi, T., and Shum, A.S. 2002. Maternal diabetes increases the risk of caudal regression caused by retinoic acid. Diabetes 51: 2811-2816.

Chang, L.K., Garcia-Cardena, G., Farnebo, F., Fannon, M., Chen, E.J., Butterfield, C., Moses, M.A., Mulligan, R.C., Folkman, J., and Kaipainen, A. 2004. Dose-dependent response of FGF-2 for lymphangiogenesis. Proc. Natl. Acad. Sci. U. S. A. 101: 11658-11663.

Chitayat, D., Kalousek, D.K., and Bamforth, J.S. 1989. Lymphatic abnormalities in fetuses with posterior cervical cystic hygroma. Am. J. Med. Genet. 33: 352-356.

Christensen, E.I., Raciti, D., Reggiani, L., Verroust, P.J., and Brandli, A.W. 2008. Gene expression analysis defines the proximal tubule as the compartment for endocytic receptor-mediated uptake in the Xenopus pronephric kidney. Pflugers Arch. 456: 1163-1176.

Clagett-Dame, M. and DeLuca, H.F. 2002. The role of vitamin A in mammalian reproduction and embryonic development. Annu. Rev. Nutr. 22: 347-381.

Cleaver, O. and Krieg, P.A. 1998. VEGF mediates angioblast migration during development of the dorsal aorta in Xenopus. Development 125: 3905-3914.

Cook, P.W., Piepkorn, M., Clegg, C.H., Plowman, G.D., DeMay, J.M., Brown, J.R., and Pittelkow, M.R. 1997. Transgenic expression of the human amphiregulin gene induces a psoriasis-like phenotype. J. Clin. Invest. 100: 2286-2294.

Cueni, L.N. and Detmar, M. 2006. New insights into the molecular control of the lymphatic vascular system and its role in disease. J. Invest. Dermatol. 126: 2167-2177.

Cueni, L.N. and Detmar, M. 2008. The lymphatic system in health and disease. Lymphat. Res. Biol. 6: 109-122.

Czyz, J. and Wobus, A. 2001. Embryonic stem cell differentiation: the role of extracellular factors. Differentiation 68: 167-174.

Dadras, S.S., Lange-Asschenfeldt, B., Velasco, P., Nguyen, L., Vora, A., Muzikansky, A., Jahnke, K., Hauschild, A., Hirakawa, S., Mihm, M.C. et al. 2005. Tumor lymphangiogenesis predicts melanoma metastasis to sentinel lymph nodes. Mod. Pathol. 18: 1232-1242.

Dang, S.M., Kyba, M., Perlingeiro, R., Daley, G.Q., and Zandstra, P.W. 2002. Efficiency of embryoid body formation and hematopoietic development from embryonic stem cells in different culture systems. Biotechnol. Bioeng. 78: 442-453.

Danussi, C., Spessotto, P., Petrucco, A., Wassermann, B., Sabatelli, P., Montesi, M., Doliana, R., Bressan, G.M., and Colombatti, A. 2008. Emilin1 deficiency causes structural and functional defects of lymphatic vasculature. Mol. Cell. Biol. 28: 4026-4039.

Dawid, I.B. and Sargent, T.D. 1988. Xenopus laevis in developmental and molecular biology. Science 240: 1443-1448.

Debes, G.F., Arnold, C.N., Young, A.J., Krautwald, S., Lipp, M., Hay, J.B., and Butcher, E.C. 2005. Chemokine receptor CCR7 required for T lymphocyte exit from peripheral tissues. Nat. Immunol. 6: 889-894.

Detmar, M., Brown, L.F., Claffey, K.P., Yeo, K.T., Kocher, O., Jackman, R.W., Berse, B., and Dvorak, H.F. 1994. Overexpression of vascular permeability factor/vascular endothelial growth factor and its receptors in psoriasis. J. Exp. Med. 180: 1141-1146.

DiGiovanna, J.J. 2001. Systemic retinoid therapy. Dermatol. Clin. 19: 161-167. Drab, M., Haller, H., Bychkov, R., Erdmann, B., Lindschau, C., Haase, H., Morano, I., Luft, F.C.,

and Wobus, A.M. 1997. From totipotent embryonic stem cells to spontaneously contracting smooth muscle cells: a retinoic acid and db-cAMP in vitro differentiation model. FASEB J. 11: 905-915.

Page 112: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

111

Dudas, J., Elmaouhoub, A., Mansuroglu, T., Batusic, D., Tron, K., Saile, B., Papoutsi, M., Pieler, T., Wilting, J., and Ramadori, G. 2006. Prospero-related homeobox 1 (Prox1) is a stable hepatocyte marker during liver development, injury and regeneration, and is absent from "oval cells". Histochem. Cell Biol. 126: 549-562.

Duester, G. 2008. Retinoic acid synthesis and signaling during early organogenesis. Cell 134: 921-931.

Dumont, D.J., Jussila, L., Taipale, J., Lymboussaki, A., Mustonen, T., Pajusola, K., Breitman, M., and Alitalo, K. 1998. Cardiovascular failure in mouse embryos deficient in VEGF receptor-3. Science 282: 946-949.

Dvash, T., Mayshar, Y., Darr, H., McElhaney, M., Barker, D., Yanuka, O., Kotkow, K.J., Rubin, L.L., Benvenisty, N., and Eiges, R. 2004. Temporal gene expression during differentiation of human embryonic stem cells and embryoid bodies. Hum. Reprod. 19: 2875-2883.

Dvorak, H.F., Detmar, M., Claffey, K.P., Nagy, J.A., van de Water, L., and Senger, D.R. 1995. Vascular permeability factor/vascular endothelial growth factor: an important mediator of angiogenesis in malignancy and inflammation. Int. Arch. Allergy Immunol. 107: 233-235.

Dyer, M.A. 2003. Regulation of proliferation, cell fate specification and differentiation by the homeodomain proteins Prox1, Six3, and Chx10 in the developing retina. Cell Cycle 2: 350-357.

Eales, N.B. 1974. The history of the lymphatic system, with special reference to the Hunter-Monro controversy. J. Hist. Med. Allied Sci. 29: 280-294.

Ebata, N., Nodasaka, Y., Sawa, Y., Yamaoka, Y., Makino, S., Totsuka, Y., and Yoshida, S. 2001. Desmoplakin as a specific marker of lymphatic vessels. Microvasc. Res. 61: 40-48.

Erhard, H., Rietveld, F.J., Brocker, E.B., de Waal, R.M., and Ruiter, D.J. 1996. Phenotype of normal cutaneous microvasculature. Immunoelectron microscopic observations with emphasis on the differences between blood vessels and lymphatics. J. Invest. Dermatol. 106: 135-140.

Evans, M.J. and Kaufman, M.H. 1981. Establishment in culture of pluripotential cells from mouse embryos. Nature 292: 154-156.

Fang, J., Dagenais, S.L., Erickson, R.P., Arlt, M.F., Glynn, M.W., Gorski, J.L., Seaver, L.H., and Glover, T.W. 2000. Mutations in FOXC2 (MFH-1), a forkhead family transcription factor, are responsible for the hereditary lymphedema-distichiasis syndrome. Am. J. Hum. Genet. 67: 1382-1388.

Ferreira, L.V., Souza, S.C., Montenegro, L.R., Malaquias, A.C., Arnhold, I.J., Mendonca, B.B., and Jorge, A.A. 2008. Analysis of the PTPN11 gene in idiopathic short stature children and Noonan syndrome patients. Clin. Endocrinol. (Oxf). 69: 426-431.

Finegold, D.N., Kimak, M.A., Lawrence, E.C., Levinson, K.L., Cherniske, E.M., Pober, B.R., Dunlap, J.W., and Ferrell, R.E. 2001. Truncating mutations in FOXC2 cause multiple lymphedema syndromes. Hum. Mol. Genet. 10: 1185-1189.

Francois, M., Caprini, A., Hosking, B., Orsenigo, F., Wilhelm, D., Browne, C., Paavonen, K., Karnezis, T., Shayan, R., Downes, M. et al. 2008. Sox18 induces development of the lymphatic vasculature in mice. Nature 456: 643-647.

Fritz-Six, K.L., Dunworth, W.P., Li, M., and Caron, K.M. 2008. Adrenomedullin signaling is necessary for murine lymphatic vascular development. J. Clin. Invest. 118: 40-50.

Fu, J., Gerhardt, H., McDaniel, J.M., Xia, B., Liu, X., Ivanciu, L., Ny, A., Hermans, K., Silasi-Mansat, R., McGee, S. et al. 2008. Endothelial cell O-glycan deficiency causes blood/lymphatic misconnections and consequent fatty liver disease in mice. J. Clin. Invest. 118: 3725-3737.

Gaetano, C., Catalano, A., Illi, B., Felici, A., Minucci, S., Palumbo, R., Facchiano, F., Mangoni, A., Mancarella, S., Muhlhauser, J. et al. 2001. Retinoids induce fibroblast growth factor-2 production in endothelial cells via retinoic acid receptor alpha activation and stimulate angiogenesis in vitro and in vivo. Circ. Res. 88: E38-47.

Page 113: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

112

Gaillard, E., Bruck, N., Brelivet, Y., Bour, G., Lalevee, S., Bauer, A., Poch, O., Moras, D., and Rochette-Egly, C. 2006. Phosphorylation by PKA potentiates retinoic acid receptor alpha activity by means of increasing interaction with and phosphorylation by cyclin H/cdk7. Proc. Natl. Acad. Sci. U. S. A. 103: 9548-9553.

Gale, N.W., Prevo, R., Espinosa, J., Ferguson, D.J., Dominguez, M.G., Yancopoulos, G.D., Thurston, G., and Jackson, D.G. 2007. Normal lymphatic development and function in mice deficient for the lymphatic hyaluronan receptor LYVE-1. Mol. Cell. Biol. 27: 595-604.

Gale, N.W., Thurston, G., Hackett, S.F., Renard, R., Wang, Q., McClain, J., Martin, C., Witte, C., Witte, M.H., Jackson, D. et al. 2002. Angiopoietin-2 is required for postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1. Dev. Cell 3: 411-423.

Gerecht, S., Burdick, J.A., Ferreira, L.S., Townsend, S.A., Langer, R., and Vunjak-Novakovic, G. 2007. Hyaluronic acid hydrogel for controlled self-renewal and differentiation of human embryonic stem cells. Proc. Natl. Acad. Sci. U. S. A. 104: 11298-11303.

Gerli, R., Ibba, L., and Fruschelli, C. 1991. Ultrastructural cytochemistry of anchoring filaments of human lymphatic capillaries and their relation to elastic fibers. Lymphology 24: 105-112.

Goldman, J., Rutkowski, J.M., Shields, J.D., Pasquier, M.C., Cui, Y., Schmokel, H.G., Willey, S., Hicklin, D.J., Pytowski, B., and Swartz, M.A. 2007. Cooperative and redundant roles of VEGFR-2 and VEGFR-3 signaling in adult lymphangiogenesis. FASEB J. 21: 1003-1012.

Grunewald, M., Avraham, I., Dor, Y., Bachar-Lustig, E., Itin, A., Jung, S., Chimenti, S., Landsman, L., Abramovitch, R., and Keshet, E. 2006. VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells. Cell 124: 175-189.

Gunn, M.D., Tangemann, K., Tam, C., Cyster, J.G., Rosen, S.D., and Williams, L.T. 1998. A chemokine expressed in lymphoid high endothelial venules promotes the adhesion and chemotaxis of naive T lymphocytes. Proc. Natl. Acad. Sci. U. S. A. 95: 258-263.

Gupta, S., Ramjaun, A.R., Haiko, P., Wang, Y., Warne, P.H., Nicke, B., Nye, E., Stamp, G., Alitalo, K., and Downward, J. 2007. Binding of ras to phosphoinositide 3-kinase p110alpha is required for ras-driven tumorigenesis in mice. Cell 129: 957-968.

Haiko, P., Makinen, T., Keskitalo, S., Taipale, J., Karkkainen, M.J., Baldwin, M.E., Stacker, S.A., Achen, M.G., and Alitalo, K. 2008. Deletion of vascular endothelial growth factor C (VEGF-C) and VEGF-D is not equivalent to VEGF receptor 3 deletion in mouse embryos. Mol. Cell. Biol. 28: 4843-4850.

Hamada, K., Oike, Y., Takakura, N., Ito, Y., Jussila, L., Dumont, D.J., Alitalo, K., and Suda, T. 2000. VEGF-C signaling pathways through VEGFR-2 and VEGFR-3 in vasculoangiogenesis and hematopoiesis. Blood 96: 3793-3800.

Harvey, N.L., Srinivasan, R.S., Dillard, M.E., Johnson, N.C., Witte, M.H., Boyd, K., Sleeman, M.W., and Oliver, G. 2005. Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity. Nat. Genet. 37: 1072-1081.

Hatzopoulos, A.K., Folkman, J., Vasile, E., Eiselen, G.K., and Rosenberg, R.D. 1998. Isolation and characterization of endothelial progenitor cells from mouse embryos. Development 125: 1457-1468.

He, Y., Karpanen, T., and Alitalo, K. 2004. Role of lymphangiogenic factors in tumor metastasis. Biochim. Biophys. Acta 1654: 3-12.

He, Y., Kozaki, K., Karpanen, T., Koshikawa, K., Yla-Herttuala, S., Takahashi, T., and Alitalo, K. 2002. Suppression of tumor lymphangiogenesis and lymph node metastasis by blocking vascular endothelial growth factor receptor 3 signaling. J. Natl. Cancer Inst. 94: 819-825.

He, Y., Rajantie, I., Pajusola, K., Jeltsch, M., Holopainen, T., Yla-Herttuala, S., Harding, T., Jooss, K., Takahashi, T., and Alitalo, K. 2005. Vascular endothelial cell growth factor receptor 3-mediated activation of lymphatic endothelium is crucial for tumor cell entry and spread via lymphatic vessels. Cancer Res. 65: 4739-4746.

Page 114: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

113

Helbling, P.M., Saulnier, D.M., and Brandli, A.W. 2000. The receptor tyrosine kinase EphB4 and ephrin-B ligands restrict angiogenic growth of embryonic veins in Xenopus laevis. Development 127: 269-278.

Helbling, P.M., Saulnier, D.M., Robinson, V., Christiansen, J.H., Wilkinson, D.G., and Brandli, A.W. 1999. Comparative analysis of embryonic gene expression defines potential interaction sites for Xenopus EphB4 receptors with ephrin-B ligands. Dev. Dyn. 216: 361-373.

Helbling, P.M., Tran, C.T., and Brandli, A.W. 1998. Requirement for EphA receptor signaling in the segregation of Xenopus third and fourth arch neural crest cells. Mech. Dev. 78: 63-79.

Herbst, R.S. and Sandler, A. 2008. Bevacizumab and erlotinib: a promising new approach to the treatment of advanced NSCLC. Oncologist 13: 1166-1176.

Hirakawa, S., Brown, L.F., Kodama, S., Paavonen, K., Alitalo, K., and Detmar, M. 2007. VEGF-C-induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites. Blood 109: 1010-1017.

Hirakawa, S., Hong, Y.K., Harvey, N., Schacht, V., Matsuda, K., Libermann, T., and Detmar, M. 2003. Identification of vascular lineage-specific genes by transcriptional profiling of isolated blood vascular and lymphatic endothelial cells. Am. J. Pathol. 162: 575-586.

Hirakawa, S., Kodama, S., Kunstfeld, R., Kajiya, K., Brown, L.F., and Detmar, M. 2005. VEGF-A induces tumor and sentinel lymph node lymphangiogenesis and promotes lymphatic metastasis. J. Exp. Med. 201: 1089-1099.

Hirashima, M., Sano, K., Morisada, T., Murakami, K., Rossant, J., and Suda, T. 2008. Lymphatic vessel assembly is impaired in Aspp1-deficient mouse embryos. Dev. Biol. 316: 149-159.

Hogan, B.M., Bos, F.L., Bussmann, J., Witte, M., Chi, N.C., Duckers, H.J., and Schulte-Merker, S. 2009. Ccbe1 is required for embryonic lymphangiogenesis and venous sprouting. Nat. Genet. 41: 396-398.

Hong, Y.K., Harvey, N., Noh, Y.H., Schacht, V., Hirakawa, S., Detmar, M., and Oliver, G. 2002. Prox1 is a master control gene in the program specifying lymphatic endothelial cell fate. Dev. Dyn. 225: 351-357.

Hong, Y.K., Lange-Asschenfeldt, B., Velasco, P., Hirakawa, S., Kunstfeld, R., Brown, L.F., Bohlen, P., Senger, D.R., and Detmar, M. 2004. VEGF-A promotes tissue repair-associated lymphatic vessel formation via VEGFR-2 and the alpha1beta1 and alpha2beta1 integrins. FASEB J. 18: 1111-1113.

Hopfl, G., Gassmann, M., and Desbaillets, I. 2004. Differentiating embryonic stem cells into embryoid bodies. Methods Mol. Biol. 254: 79-98.

Hoshida, T., Isaka, N., Hagendoorn, J., di Tomaso, E., Chen, Y.L., Pytowski, B., Fukumura, D., Padera, T.P., and Jain, R.K. 2006. Imaging steps of lymphatic metastasis reveals that vascular endothelial growth factor-C increases metastasis by increasing delivery of cancer cells to lymph nodes: therapeutic implications. Cancer Res. 66: 8065-8075.

Huang, S.S., Liu, I.H., Smith, T., Shah, M.R., Johnson, F.E., and Huang, J.S. 2006. CRSBP-1/LYVE-l-null mice exhibit identifiable morphological and functional alterations of lymphatic capillary vessels. FEBS Lett. 580: 6259-6268.

Huang, X.Z., Wu, J.F., Ferrando, R., Lee, J.H., Wang, Y.L., Farese, R.V., Jr., and Sheppard, D. 2000. Fatal bilateral chylothorax in mice lacking the integrin alpha9beta1. Mol. Cell. Biol. 20: 5208-5215.

Huntington, G.S. and McClure, C.F.W. 1910. The anatomy and development of the jugular lymph sac in the domestic cat (Felis domestica). Am. J. Anat. 10: 177-312.

Ichise, H., Ichise, T., Ohtani, O., and Yoshida, N. 2009. Phospholipase Cgamma2 is necessary for separation of blood and lymphatic vasculature in mice. Development 136: 191-195.

Irjala, H., Alanen, K., Grenman, R., Heikkila, P., Joensuu, H., and Jalkanen, S. 2003. Mannose receptor (MR) and common lymphatic endothelial and vascular endothelial receptor

Page 115: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

114

(CLEVER)-1 direct the binding of cancer cells to the lymph vessel endothelium. Cancer Res. 63: 4671-4676.

Irjala, H., Johansson, E.L., Grenman, R., Alanen, K., Salmi, M., and Jalkanen, S. 2001. Mannose receptor is a novel ligand for L-selectin and mediates lymphocyte binding to lymphatic endothelium. J. Exp. Med. 194: 1033-1042.

Irrthum, A., Devriendt, K., Chitayat, D., Matthijs, G., Glade, C., Steijlen, P.M., Fryns, J.P., Van Steensel, M.A., and Vikkula, M. 2003. Mutations in the transcription factor gene SOX18 underlie recessive and dominant forms of hypotrichosis-lymphedema-telangiectasia. Am. J. Hum. Genet. 72: 1470-1478.

Itskovitz-Eldor, J., Schuldiner, M., Karsenti, D., Eden, A., Yanuka, O., Amit, M., Soreq, H., and Benvenisty, N. 2000. Differentiation of human embryonic stem cells into embryoid bodies compromising the three embryonic germ layers. Mol. Med. 6: 88-95.

Jackson, D.G., Prevo, R., Clasper, S., and Banerji, S. 2001. LYVE-1, the lymphatic system and tumor lymphangiogenesis. Trends Immunol. 22: 317-321.

Jeltsch, M., Kaipainen, A., Joukov, V., Meng, X., Lakso, M., Rauvala, H., Swartz, M., Fukumura, D., Jain, R.K., and Alitalo, K. 1997. Hyperplasia of lymphatic vessels in VEGF-C transgenic mice. Science 276: 1423-1425.

Jin, D., Harada, K., Ohnishi, S., Yamahara, K., Kangawa, K., and Nagaya, N. 2008. Adrenomedullin induces lymphangiogenesis and ameliorates secondary lymphoedema. Cardiovasc. Res. 80: 339-345.

Joukov, V., Kumar, V., Sorsa, T., Arighi, E., Weich, H., Saksela, O., and Alitalo, K. 1998. A recombinant mutant vascular endothelial growth factor-C that has lost vascular endothelial growth factor receptor-2 binding, activation, and vascular permeability activities. J. Biol. Chem. 273: 6599-6602.

Joukov, V., Sorsa, T., Kumar, V., Jeltsch, M., Claesson-Welsh, L., Cao, Y., Saksela, O., Kalkkinen, N., and Alitalo, K. 1997. Proteolytic processing regulates receptor specificity and activity of VEGF-C. EMBO J. 16: 3898-3911.

Jurisic, G. and Detmar, M. 2009. Lymphatic endothelium in health and disease. Cell Tissue Res. 335: 97-108.

Kaipainen, A., Korhonen, J., Mustonen, T., van Hinsbergh, V.W., Fang, G.H., Dumont, D., Breitman, M., and Alitalo, K. 1995. Expression of the fms-like tyrosine kinase 4 gene becomes restricted to lymphatic endothelium during development. Proc. Natl. Acad. Sci. U. S. A. 92: 3566-3570.

Kajiya, K., Hirakawa, S., and Detmar, M. 2006. Vascular endothelial growth factor-A mediates ultraviolet B-induced impairment of lymphatic vessel function. Am. J. Pathol. 169: 1496-1503.

Kajiya, K., Hirakawa, S., Ma, B., Drinnenberg, I., and Detmar, M. 2005. Hepatocyte growth factor promotes lymphatic vessel formation and function. EMBO J. 24: 2885-2895.

Kalin, R.E., Banziger-Tobler, N.E., Detmar, M., and Brandli, A.W. 2009. An in vivo chemical library screen in Xenopus tadpoles reveals novel pathways involved in angiogenesis and lymphangiogenesis. Blood 114: 1110-1122

Kalin, R.E., Kretz, M.P., Meyer, A.M., Kispert, A., Heppner, F.L., and Brandli, A.W. 2007. Paracrine and autocrine mechanisms of apelin signaling govern embryonic and tumor angiogenesis. Dev. Biol. 305: 599-614.

Karkkainen, M.J., Haiko, P., Sainio, K., Partanen, J., Taipale, J., Petrova, T.V., Jeltsch, M., Jackson, D.G., Talikka, M., Rauvala, H. et al. 2004. Vascular endothelial growth factor C is required for sprouting of the first lymphatic vessels from embryonic veins. Nat. Immunol. 5: 74-80.

Karkkainen, M.J., Jussila, L., Ferrell, R.E., Finegold, D.N., and Alitalo, K. 2001. Molecular regulation of lymphangiogenesis and targets for tissue oedema. Trends Mol. Med. 7: 18-22.

Page 116: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

115

Karpanen, T. and Alitalo, K. 2008. Molecular biology and pathology of lymphangiogenesis. Annu. Rev. Pathol. 3: 367-397.

Karpanen, T., Egeblad, M., Karkkainen, M.J., Kubo, H., Yla-Herttuala, S., Jaattela, M., and Alitalo, K. 2001. Vascular endothelial growth factor C promotes tumor lymphangiogenesis and intralymphatic tumor growth. Cancer Res. 61: 1786-1790.

Karpanen, T., Heckman, C.A., Keskitalo, S., Jeltsch, M., Ollila, H., Neufeld, G., Tamagnone, L., and Alitalo, K. 2006. Functional interaction of VEGF-C and VEGF-D with neuropilin receptors. FASEB J. 20: 1462-1472.

Kato, Y., Fujita, N., Kunita, A., Sato, S., Kaneko, M., Osawa, M., and Tsuruo, T. 2003. Molecular identification of Aggrus/T1alpha as a platelet aggregation-inducing factor expressed in colorectal tumors. J. Biol. Chem. 278: 51599-51605.

Kerjaschki, D. 2005. The crucial role of macrophages in lymphangiogenesis. J. Clin. Invest. 115: 2316-2319.

Kerjaschki, D. 2006. Lymphatic neoangiogenesis in renal transplants: a driving force of chronic rejection? J. Nephrol. 19: 403-406.

Kerjaschki, D., Regele, H.M., Moosberger, I., Nagy-Bojarski, K., Watschinger, B., Soleiman, A., Birner, P., Krieger, S., Hovorka, A., Silberhumer, G. et al. 2004. Lymphatic neoangiogenesis in human kidney transplants is associated with immunologically active lymphocytic infiltrates. J. Am. Soc. Nephrol. 15: 603-612.

Kochhar, D.M. 1967. Teratogenetic activity of retinoic acid. Acta Pathol. Microbiol. Scand. 70: 398-404.

Koide, T., Downes, M., Chandraratna, R.A., Blumberg, B., and Umesono, K. 2001. Active repression of RAR signaling is required for head formation. Genes Dev. 15: 2111-2121.

Kreuger, J., Nilsson, I., Kerjaschki, D., Petrova, T., Alitalo, K., and Claesson-Welsh, L. 2006. Early lymph vessel development from embryonic stem cells. Arterioscler. Thromb. Vasc. Biol. 26: 1073-1078.

Kriehuber, E., Breiteneder-Geleff, S., Groeger, M., Soleiman, A., Schoppmann, S.F., Stingl, G., Kerjaschki, D., and Maurer, D. 2001. Isolation and characterization of dermal lymphatic and blood endothelial cells reveal stable and functionally specialized cell lineages. J. Exp. Med. 194: 797-808.

Kubo, H., Cao, R., Brakenhielm, E., Makinen, T., Cao, Y., and Alitalo, K. 2002. Blockade of vascular endothelial growth factor receptor-3 signaling inhibits fibroblast growth factor-2-induced lymphangiogenesis in mouse cornea. Proc. Natl. Acad. Sci. U. S. A. 99: 8868-8873.

Kulkarni, R.M., Greenberg, J.M., and Akeson, A.L. 2009. NFATc1 regulates lymphatic endothelial development. Mech. Dev. 126: 350-365.

Kumar, R. and Yarmand-Bagheri, R. 2001. The role of HER2 in angiogenesis. Semin. Oncol. 28: 27-32.

Kunstfeld, R., Hirakawa, S., Hong, Y.K., Schacht, V., Lange-Asschenfeldt, B., Velasco, P., Lin, C., Fiebiger, E., Wei, X., Wu, Y. et al. 2004. Induction of cutaneous delayed-type hypersensitivity reactions in VEGF-A transgenic mice results in chronic skin inflammation associated with persistent lymphatic hyperplasia. Blood 104: 1048-1057.

Kurosawa, H. 2007. Methods for inducing embryoid body formation: in vitro differentiation system of embryonic stem cells. J . Biosci. Bioeng. 103: 389-398.

Kyzas, P.A., Geleff, S., Batistatou, A., Agnantis, N.J., and Stefanou, D. 2005. Evidence for lymphangiogenesis and its prognostic implications in head and neck squamous cell carcinoma. J. Pathol. 206: 170-177.

Lahdenranta, J., Hagendoorn, J., Padera, T.P., Hoshida, T., Nelson, G., Kashiwagi, S., Jain, R.K., and Fukumura, D. 2009. Endothelial nitric oxide synthase mediates lymphangiogenesis and lymphatic metastasis. Cancer Res. 69: 2801-2808.

Lammer, E., Chen, D., Hoar, R., Agnish, N., Benke, P., Braun, J., Curry, C., Fernhoff, P., Grix, A., and Lott, I. 1986. Retinoic acid embryopathy. N. Engl. J. Med. 315: 262-263.

Page 117: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

116

Lang, E., Mazauric-Stuker, M.L., and Maelicke, A. 1989. States of developmental commitment of a mouse embryonal carcinoma cell line differentiating along a neural pathway. J. Cell Biol. 109: 2481-2493.

Leahy, A., Xiong, J.W., Kuhnert, F., and Stuhlmann, H. 1999. Use of developmental marker genes to define temporal and spatial patterns of differentiation during embryoid body formation. J. Exp. Zool. 284: 67-81.

Leak, L.V. and Burke, J.F. 1968. Electron microscopic study of lymphatic capillaries in the removal of connective tissue fluids and particulate substances. Lymphology 1: 39-52.

Lee, S., Kang, J., Yoo, J., Ganesan, S.K., Cook, S.C., Aguilar, B., Ramu, S., Lee, J., and Hong, Y.K. 2009. Prox1 physically and functionally interacts with COUP-TFII to specify lymphatic endothelial cell fate. Blood 113: 1856-1859.

Leelasiri, A., Numbenjapol, T., Prayoonwiwat, W., Mongkolsritrakul, W., and Srisawat, C. 2005. Successful treatment of retinoic acid syndrome with dexamethasone: a case report. J. Med. Assoc. Thai. 88: S302-310.

Levine, A.J., Munoz-Sanjuan, I., Bell, E., North, A.J., and Brivanlou, A.H. 2003. Fluorescent labeling of endothelial cells allows in vivo, continuous characterization of the vascular development of Xenopus laevis. Dev. Biol. 254: 50-67.

Liersch, R., Nay, F., Lu, L., and Detmar, M. 2006. Induction of lymphatic endothelial cell differentiation in embryoid bodies. Blood 107: 1214-1216.

Lin, J., Lalani, A.S., Harding, T.C., Gonzalez, M., Wu, W.W., Luan, B., Tu, G.H., Koprivnikar, K., VanRoey, M.J., He, Y. et al. 2005. Inhibition of lymphogenous metastasis using adeno-associated virus-mediated gene transfer of a soluble VEGFR-3 decoy receptor. Cancer Res. 65: 6901-6909.

Lingen, M.W., Polverini, P.J., and Bouck, N.P. 1996. Inhibition of squamous cell carcinoma angiogenesis by direct interaction of retinoic acid with endothelial cells. Lab. Invest. 74: 476-483.

Liu, N.F. and He, Q.L. 1997. The regulatory effects of cytokines on lymphatic angiogenesis. Lymphology 30: 3-12.

Lohnes, D., Mark, M., Mendelsohn, C., Dolle, P., Decimo, D., LeMeur, M., Dierich, A., Gorry, P., and Chambon, P. 1995. Developmental roles of the retinoic acid receptors. J. Steroid Biochem. Mol. Biol. 53: 475-486.

Lord, R.S. 1968. The white veins: conceptual difficulties in the history of the lymphatics. Med. Hist. 12: 174-184.

Machnik, A., Neuhofer, W., Jantsch, J., Dahlmann, A., Tammela, T., Machura, K., Park, J.K., Beck, F.X., Muller, D.N., Derer, W. et al. 2009. Macrophages regulate salt-dependent volume and blood pressure by a vascular endothelial growth factor-C-dependent buffering mechanism. Nat. Med. 15: 545-552.

Majewski, S., Marczak, M., Szmurlo, A., Jablonska, S., and Bollag, W. 1995. Retinoids, interferon alpha, 1,25-dihydroxyvitamin D3 and their combination inhibit angiogenesis induced by non-HPV-harboring tumor cell lines. RAR alpha mediates the antiangiogenic effect of retinoids. Cancer Lett. 89: 117-124.

Makinen, T., Adams, R.H., Bailey, J., Lu, Q., Ziemiecki, A., Alitalo, K., Klein, R., and Wilkinson, G.A. 2005. PDZ interaction site in ephrinB2 is required for the remodeling of lymphatic vasculature. Genes Dev. 19: 397-410.

Makinen, T. and Alitalo, K. 2002. Molecular mechanisms of lymphangiogenesis. Cold Spring Harb. Symp. Quant. Biol. 67: 189-196.

Makinen, T., Jussila, L., Veikkola, T., Karpanen, T., Kettunen, M.I., Pulkkanen, K.J., Kauppinen, R., Jackson, D.G., Kubo, H., Nishikawa, S. et al. 2001. Inhibition of lymphangiogenesis with resulting lymphedema in transgenic mice expressing soluble VEGF receptor-3. Nat. Med. 7: 199-205.

Page 118: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

117

Malaquias, A.C., Ferreira, L.V., Souza, S.C., Arnhold, I.J., Mendonca, B.B., and Jorge, A.A. 2008. Noonan syndrome: from phenotype to growth hormone therapy. Arq. Bras. Endocrinol. Metabol. 52: 800-808.

Mandala, S., Hajdu, R., Bergstrom, J., Quackenbush, E., Xie, J., Milligan, J., Thornton, R., Shei, G.J., Card, D., Keohane, C. et al. 2002. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science 296: 346-349.

Mandriota, S.J., Jussila, L., Jeltsch, M., Compagni, A., Baetens, D., Prevo, R., Banerji, S., Huarte, J., Montesano, R., Jackson, D.G. et al. 2001. Vascular endothelial growth factor-C-mediated lymphangiogenesis promotes tumour metastasis. EMBO J. 20: 672-682.

Manolache, L., Benea, V., and Petrescu-Seceleanu, D. 2009. A case of solid facial oedema successfully treated with isotretinoin. J. Eur. Acad. Dermatol. Venereol. 23: 965-966.

Manshouri, T., Yang, Y., Lin, H., Stass, S.A., Glassman, A.B., Keating, M.J., and Albitar, M. 1997. Downregulation of RAR alpha in mice by antisense transgene leads to a compensatory increase in RAR beta and RAR gamma and development of lymphoma. Blood 89: 2507-2515.

Martin-Villar, E., Megias, D., Castel, S., Yurrita, M.M., Vilaro, S., and Quintanilla, M. 2006. Podoplanin binds ERM proteins to activate RhoA and promote epithelial-mesenchymal transition. J. Cell Sci. 119: 4541-4553.

Maruyama, K., Ii, M., Cursiefen, C., Jackson, D.G., Keino, H., Tomita, M., Van Rooijen, N., Takenaka, H., D'Amore, P.A., Stein-Streilein, J. et al. 2005. Inflammation-induced lymphangiogenesis in the cornea arises from CD11b-positive macrophages. J. Clin. Invest. 115: 2363-2372.

Matloubian, M., Lo, C.G., Cinamon, G., Lesneski, M.J., Xu, Y., Brinkmann, V., Allende, M.L., Proia, R.L., and Cyster, J.G. 2004. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 427: 355-360.

Melrose, W.D. 2002. Lymphatic filariasis: new insights into an old disease. Int. J. Parasitol. 32: 947-960.

Mishima, K., Watabe, T., Saito, A., Yoshimatsu, Y., Imaizumi, N., Masui, S., Hirashima, M., Morisada, T., Oike, Y., Araie, M. et al. 2007. Prox1 induces lymphatic endothelial differentiation via integrin alpha9 and other signaling cascades. Mol. Biol. Cell 18: 1421-1429.

Molotkov, A., Molotkova, N., and Duester, G. 2005. Retinoic acid generated by Raldh2 in mesoderm is required for mouse dorsal endodermal pancreas development. Dev. Dyn. 232: 950-957.

Morisada, T., Oike, Y., Yamada, Y., Urano, T., Akao, M., Kubota, Y., Maekawa, H., Kimura, Y., Ohmura, M., Miyamoto, T. et al. 2005. Angiopoietin-1 promotes LYVE-1-positive lymphatic vessel formation. Blood 105: 4649-4656.

Moro Balbas, J.A., Gato, A., Alonso Revuelta, M.I., Pastor, J.F., Repressa, J.J., and Barbosa, E. 1993. Retinoic acid induces changes in the rhombencephalic neural crest cells migration and extracellular matrix composition in chick embryos. Teratology 48: 197-206.

Mouta Carreira, C., Nasser, S.M., di Tomaso, E., Padera, T.P., Boucher, Y., Tomarev, S.I., and Jain, R.K. 2001. LYVE-1 is not restricted to the lymph vessels: expression in normal liver blood sinusoids and down-regulation in human liver cancer and cirrhosis. Cancer Res. 61: 8079-8084.

Ng, E.S., Davis, R.P., Azzola, L., Stanley, E.G., and Elefanty, A.G. 2005. Forced aggregation of defined numbers of human embryonic stem cells into embryoid bodies fosters robust, reproducible hematopoietic differentiation. Blood 106: 1601-1603.

Nonaka, H., Tanaka, M., Suzuki, K., and Miyajima, A. 2007. Development of murine hepatic sinusoidal endothelial cells characterized by the expression of hyaluronan receptors. Dev. Dyn. 236: 2258-2267.

Page 119: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

118

Norrmen, C., Ivanov, K.I., Cheng, J., Zangger, N., Delorenzi, M., Jaquet, M., Miura, N., Puolakkainen, P., Horsley, V., Hu, J. et al. 2009. FOXC2 controls formation and maturation of lymphatic collecting vessels through cooperation with NFATc1. J. Cell Biol. 185: 439-457.

Ny, A., Koch, M., Schneider, M., Neven, E., Tong, R.T., Maity, S., Fischer, C., Plaisance, S., Lambrechts, D., Heligon, C. et al. 2005. A genetic Xenopus laevis tadpole model to study lymphangiogenesis. Nat. Med. 11: 998-1004.

Ny, A., Koch, M., Vandevelde, W., Schneider, M., Fischer, C., Diez-Juan, A., Neven, E., Geudens, I., Maity, S., Moons, L. et al. 2008. Role of VEGF-D and VEGFR-3 in developmental lymphangiogenesis, a chemicogenetic study in Xenopus tadpoles. Blood 112: 1740-1749.

O'Shea, K.S. 2004. Self-renewal vs. differentiation of mouse embryonic stem cells. Biol. Reprod. 71: 1755-1765.

Ogura, Y., Suruga, K., Mochizuki, H., Yamamoto, T., Takase, S., and Goda, T. 2004. Postnatal changes in gene expression of retinal dehydrogenase and retinoid receptors in liver of rats. Life Sci. 74: 1519-1528.

Ohl, L., Mohaupt, M., Czeloth, N., Hintzen, G., Kiafard, Z., Zwirner, J., Blankenstein, T., Henning, G., and Forster, R. 2004. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 21: 279-288.

Oikawa, T., Hirotani, K., Nakamura, O., Shudo, K., Hiragun, A., and Iwaguchi, T. 1989. A highly potent antiangiogenic activity of retinoids. Cancer Lett. 48: 157-162.

Oka, M., Iwata, C., Suzuki, H.I., Kiyono, K., Morishita, Y., Watabe, T., Komuro, A., Kano, M.R., and Miyazono, K. 2008. Inhibition of endogenous TGF-beta signaling enhances lymphangiogenesis. Blood 111: 4571-4579.

Oliver, G. 2004. Lymphatic vasculature development. Nat. Rev. Immunol. 4: 35-45. Oliver, G. and Detmar, M. 2002. The rediscovery of the lymphatic system: old and new insights

into the development and biological function of the lymphatic vasculature. Genes Dev. 16: 773-783.

Orfanos, C.E., Zouboulis, C.C., Almond-Roesler, B., and Geilen, C.C. 1997. Current use and future potential role of retinoids in dermatology. Drugs 53: 358-388.

Ozeki, H. and Shirai, S. 1998. Developmental eye abnormalities in mouse fetuses induced by retinoic acid. Jpn. J. Ophthalmol. 42: 162-167.

Paavonen, K., Mandelin, J., Partanen, T., Jussila, L., Li, T.F., Ristimaki, A., Alitalo, K., and Konttinen, Y.T. 2002. Vascular endothelial growth factors C and D and their VEGFR-2 and 3 receptors in blood and lymphatic vessels in healthy and arthritic synovium. J. Rheumatol. 29: 39-45.

Pepper, M.S., Tille, J.C., Nisato, R., and Skobe, M. 2003. Lymphangiogenesis and tumor metastasis. Cell Tissue Res. 314: 167-177.

Perez-Soler, R. and Saltz, L. 2005. Cutaneous adverse effects with HER1/EGFR-targeted agents: is there a silver lining? J. Clin. Oncol. 23: 5235-5246.

Perrotte, P., Matsumoto, T., Inoue, K., Kuniyasu, H., Eve, B.Y., Hicklin, D.J., Radinsky, R., and Dinney, C.P. 1999. Anti-epidermal growth factor receptor antibody C225 inhibits angiogenesis in human transitional cell carcinoma growing orthotopically in nude mice. Clin. Cancer Res. 5: 257-265.

Petit, A.M., Rak, J., Hung, M.C., Rockwell, P., Goldstein, N., Fendly, B., and Kerbel, R.S. 1997. Neutralizing antibodies against epidermal growth factor and ErbB-2/neu receptor tyrosine kinases down-regulate vascular endothelial growth factor production by tumor cells in vitro and in vivo: angiogenic implications for signal transduction therapy of solid tumors. Am. J. Pathol. 151: 1523-1530.

Petrova, T.V., Karpanen, T., Norrmen, C., Mellor, R., Tamakoshi, T., Finegold, D., Ferrell, R., Kerjaschki, D., Mortimer, P., Yla-Herttuala, S. et al. 2004. Defective valves and abnormal

Page 120: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

119

mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis. Nat. Med. 10: 974-981.

Petrova, T.V., Makinen, T., Makela, T.P., Saarela, J., Virtanen, I., Ferrell, R.E., Finegold, D.N., Kerjaschki, D., Yla-Herttuala, S., and Alitalo, K. 2002. Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor. EMBO J. 21: 4593-4599.

Pino-Lagos, K., Benson, M.J., and Noelle, R.J. 2008. Retinoic acid in the immune system. Ann. N. Y. Acad. Sci. 1143: 170-187.

Podgrabinska, S., Braun, P., Velasco, P., Kloos, B., Pepper, M.S., and Skobe, M. 2002. Molecular characterization of lymphatic endothelial cells. Proc. Natl. Acad. Sci. U. S. A. 99: 16069-16074.

Prevo, R., Banerji, S., Ferguson, D.J., Clasper, S., and Jackson, D.G. 2001. Mouse LYVE-1 is an endocytic receptor for hyaluronan in lymphatic endothelium. J. Biol. Chem. 276: 19420-19430.

Purhonen, S., Palm, J., Rossi, D., Kaskenpaa, N., Rajantie, I., Yla-Herttuala, S., Alitalo, K., Weissman, I.L., and Salven, P. 2008. Bone marrow-derived circulating endothelial precursors do not contribute to vascular endothelium and are not needed for tumor growth. Proc. Natl. Acad. Sci. U. S. A. 105: 6620-6625.

Quemelo, P.R., Lourenco, C.M., and Peres, L.C. 2007. Teratogenic effect of retinoic acid in swiss mice. Acta. Cir. Bras. 22: 451-456.

Raciti, D., Reggiani, L., Geffers, L., Jiang, Q., Bacchion, F., Subrizi, A.E., Clements, D., Tindal, C., Davidson, D.R., Kaissling, B. et al. 2008. Organization of the pronephric kidney revealed by large-scale gene expression mapping. Genome Biol. 9: R84.

Radhika, M.S., Bhaskaram, P., Balakrishna, N., Ramalakshmi, B.A., Devi, S., and Kumar, B.S. 2002. Effects of vitamin A deficiency during pregnancy on maternal and child health. Bjog 109: 689-693.

Rebhun, R.B., Langley, R.R., Yokoi, K., Fan, D., Gershenwald, J.E., and Fidler, I.J. 2006. Targeting receptor tyrosine kinase on lymphatic endothelial cells for the therapy of colon cancer lymph node metastasis. Neoplasia 8: 747-757.

Religa, P., Cao, R., Bjorndahl, M., Zhou, Z., Zhu, Z., and Cao, Y. 2005. Presence of bone marrow-derived circulating progenitor endothelial cells in the newly formed lymphatic vessels. Blood 106: 4184-4190.

Riese, D.J., 2nd and Stern, D.F. 1998. Specificity within the EGF family/ErbB receptor family signaling network. Bioessays 20: 41-48.

Rinderknecht, M. and Detmar, M. 2008. Tumor lymphangiogenesis and melanoma metastasis. J. Cell. Physiol. 216: 347-354.

Risau, W., Sariola, H., Zerwes, H.G., Sasse, J., Ekblom, P., Kemler, R., and Doetschman, T. 1988. Vasculogenesis and angiogenesis in embryonic-stem-cell-derived embryoid bodies. Development 102: 471-478.

Ristimaki, A., Narko, K., Enholm, B., Joukov, V., and Alitalo, K. 1998. Proinflammatory cytokines regulate expression of the lymphatic endothelial mitogen vascular endothelial growth factor-C. J. Biol. Chem. 273: 8413-8418.

Roberts, N., Kloos, B., Cassella, M., Podgrabinska, S., Persaud, K., Wu, Y., Pytowski, B., and Skobe, M. 2006. Inhibition of VEGFR-3 activation with the antagonistic antibody more potently suppresses lymph node and distant metastases than inactivation of VEGFR-2. Cancer Res. 66: 2650-2657.

Rockson, S.G. 2001. Lymphedema. Am. J. Med. 110: 288-295. Saaristo, A., Karkkainen, M.J., and Alitalo, K. 2002a. Insights into the molecular pathogenesis and

targeted treatment of lymphedema. Ann. N. Y. Acad. Sci. 979: 94-110. Saaristo, A., Veikkola, T., Enholm, B., Hytonen, M., Arola, J., Pajusola, K., Turunen, P., Jeltsch,

M., Karkkainen, M.J., Kerjaschki, D. et al. 2002b. Adenoviral VEGF-C overexpression

Page 121: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

120

induces blood vessel enlargement, tortuosity, and leakiness but no sprouting angiogenesis in the skin or mucous membranes. FASEB J. 16: 1041-1049.

Sabin, F. 1902. On the origin of the lymphatics system from the veins and the development of the lymph hearts and the thoracic duct in the pig. Am. J. Anat. 1: 367-389.

Sacchi, G., Weber, E., Agliano, M., Raffaelli, N., and Comparini, L. 1997. The structure of superficial lymphatics in the human thigh: precollectors. Anat. Rec. 247: 53-62.

Saeki, H., Moore, A.M., Brown, M.J., and Hwang, S.T. 1999. Cutting edge: secondary lymphoid-tissue chemokine (SLC) and CC chemokine receptor 7 (CCR7) participate in the emigration pathway of mature dendritic cells from the skin to regional lymph nodes. J. Immunol. 162: 2472-2475.

Saito, A., Sugawara, A., Uruno, A., Kudo, M., Kagechika, H., Sato, Y., Owada, Y., Kondo, H., Sato, M., Kurabayashi, M. et al. 2007. All-trans retinoic acid induces in vitro angiogenesis via retinoic acid receptor: possible involvement of paracrine effects of endogenous vascular endothelial growth factor signaling. Endocrinology 148: 1412-1423.

Sallusto, F., Schaerli, P., Loetscher, P., Schaniel, C., Lenig, D., Mackay, C.R., Qin, S., and Lanzavecchia, A. 1998. Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation. Eur. J. Immunol. 28: 2760-2769.

Salmi, M., Koskinen, K., Henttinen, T., Elima, K., and Jalkanen, S. 2004. CLEVER-1 mediates lymphocyte transmigration through vascular and lymphatic endothelium. Blood 104: 3849-3857.

Saulnier, D.M., Ghanbari, H., and Brandli, A.W. 2002. Essential function of Wnt-4 for tubulogenesis in the Xenopus pronephric kidney. Dev. Biol. 248: 13-28.

Saurat, J.H. 1998. Systemic retinoids. What's new? Dermatol. Clin. 16: 331-340. Scavelli, C., Vacca, A., Di Pietro, G., Dammacco, F., and Ribatti, D. 2004. Crosstalk between

angiogenesis and lymphangiogenesis in tumor progression. Leukemia 18: 1054-1058. Schacht, V., Ramirez, M.I., Hong, Y.K., Hirakawa, S., Feng, D., Harvey, N., Williams, M., Dvorak,

A.M., Dvorak, H.F., Oliver, G. et al. 2003. T1alpha/podoplanin deficiency disrupts normal lymphatic vasculature formation and causes lymphedema. EMBO J. 22: 3546-3556.

Schledzewski, K., Falkowski, M., Moldenhauer, G., Metharom, P., Kzhyshkowska, J., Ganss, R., Demory, A., Falkowska-Hansen, B., Kurzen, H., Ugurel, S. et al. 2006. Lymphatic endothelium-specific hyaluronan receptor LYVE-1 is expressed by stabilin-1+, F4/80+, CD11b+ macrophages in malignant tumours and wound healing tissue in vivo and in bone marrow cultures in vitro: implications for the assessment of lymphangiogenesis. J. Pathol. 209: 67-77.

Schmid-Schonbein, G.W. 2003. The second valve system in lymphatics. Lymphat. Res. Biol. 1: 25-29.

Schoppmann, S.F., Birner, P., Stockl, J., Kalt, R., Ullrich, R., Caucig, C., Kriehuber, E., Nagy, K., Alitalo, K., and Kerjaschki, D. 2002. Tumor-associated macrophages express lymphatic endothelial growth factors and are related to peritumoral lymphangiogenesis. Am. J. Pathol. 161: 947-956.

Schreiber, A.B., Winkler, M.E., and Derynck, R. 1986. Transforming growth factor-alpha: a more potent angiogenic mediator than epidermal growth factor. Science 232: 1250-1253.

Sebzda, E., Hibbard, C., Sweeney, S., Abtahian, F., Bezman, N., Clemens, G., Maltzman, J.S., Cheng, L., Liu, F., Turner, M. et al. 2006. Syk and Slp-76 mutant mice reveal a cell-autonomous hematopoietic cell contribution to vascular development. Dev. Cell 11: 349-361.

Seo, S., Fujita, H., Nakano, A., Kang, M., Duarte, A., and Kume, T. 2006. The forkhead transcription factors, Foxc1 and Foxc2, are required for arterial specification and lymphatic sprouting during vascular development. Dev. Biol. 294: 458-470.

Shibata, M.A., Morimoto, J., Shibata, E., and Otsuki, Y. 2008. Combination therapy with short interfering RNA vectors against VEGF-C and VEGF-A suppresses lymph node and lung

Page 122: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

121

metastasis in a mouse immunocompetent mammary cancer model. Cancer Gene Ther. 15: 776-786.

Shields, J.D., Emmett, M.S., Dunn, D.B., Joory, K.D., Sage, L.M., Rigby, H., Mortimer, P.S., Orlando, A., Levick, J.R., and Bates, D.O. 2007. Chemokine-mediated migration of melanoma cells towards lymphatics--a mechanism contributing to metastasis. Oncogene 26: 2997-3005.

Shimoda, H., Bernas, M.J., Witte, M.H., Gale, N.W., Yancopoulos, G.D., and Kato, S. 2007. Abnormal recruitment of periendothelial cells to lymphatic capillaries in digestive organs of angiopoietin-2-deficient mice. Cell Tissue Res. 328: 329-337.

Shin, J.W., Min, M., Larrieu-Lahargue, F., Canron, X., Kunstfeld, R., Nguyen, L., Henderson, J.E., Bikfalvi, A., Detmar, M., and Hong, Y.K. 2006. Prox1 promotes lineage-specific expression of fibroblast growth factor (FGF) receptor-3 in lymphatic endothelium: a role for FGF signaling in lymphangiogenesis. Mol. Biol. Cell 17: 576-584.

Shudo, K., Fukasawa, H., Nakagomi, M., and Yamagata, N. 2009. Towards retinoid therapy for Alzheimer's disease. Curr. Alzheimer Res. 6: 302-311.

Sini, P., Wyder, L., Schnell, C., O'Reilly, T., Littlewood, A., Brandt, R., Hynes, N.E., and Wood, J. 2005. The antitumor and antiangiogenic activity of vascular endothelial growth factor receptor inhibition is potentiated by ErbB1 blockade. Clin. Cancer Res. 11: 4521-4532.

Skobe, M., Hawighorst, T., Jackson, D.G., Prevo, R., Janes, L., Velasco, P., Riccardi, L., Alitalo, K., Claffey, K., and Detmar, M. 2001. Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat. Med. 7: 192-198.

Slavotinek, A., Lee, S.S., Davis, R., Shrit, A., Leppig, K.A., Rhim, J., Jasnosz, K., Albertson, D., and Pinkel, D. 2005. Fryns syndrome phenotype caused by chromosome microdeletions at 15q26.2 and 8p23.1. J. Med. Genet. 42: 730-736.

Soker, S., Miao, H.Q., Nomi, M., Takashima, S., and Klagsbrun, M. 2002. VEGF165 mediates formation of complexes containing VEGFR-2 and neuropilin-1 that enhance VEGF165-receptor binding. J. Cell. Biochem. 85: 357-368.

Srinivasan, R.S., Dillard, M.E., Lagutin, O.V., Lin, F.J., Tsai, S., Tsai, M.J., Samokhvalov, I.M., and Oliver, G. 2007. Lineage tracing demonstrates the venous origin of the mammalian lymphatic vasculature. Genes Dev. 21: 2422-2432.

Stacker, S.A., Achen, M.G., Jussila, L., Baldwin, M.E., and Alitalo, K. 2002a. Lymphangiogenesis and cancer metastasis. Nat. Rev. Cancer 2: 573-583.

Stacker, S.A., Baldwin, M.E., and Achen, M.G. 2002b. The role of tumor lymphangiogenesis in metastatic spread. FASEB J. 16: 922-934.

Stacker, S.A., Caesar, C., Baldwin, M.E., Thornton, G.E., Williams, R.A., Prevo, R., Jackson, D.G., Nishikawa, S., Kubo, H., and Achen, M.G. 2001. VEGF-D promotes the metastatic spread of tumor cells via the lymphatics. Nat. Med. 7: 186-191.

Suzuki-Inoue, K., Kato, Y., Inoue, O., Kaneko, M.K., Mishima, K., Yatomi, Y., Yamazaki, Y., Narimatsu, H., and Ozaki, Y. 2007. Involvement of the snake toxin receptor CLEC-2, in podoplanin-mediated platelet activation, by cancer cells. J. Biol. Chem. 282: 25993-26001.

Szuba, A., Skobe, M., Karkkainen, M.J., Shin, W.S., Beynet, D.P., Rockson, N.B., Dakhil, N., Spilman, S., Goris, M.L., Strauss, H.W. et al. 2002. Therapeutic lymphangiogenesis with human recombinant VEGF-C. FASEB J. 16: 1985-1987.

Tammela, T., Saaristo, A., Holopainen, T., Lyytikka, J., Kotronen, A., Pitkonen, M., Abo-Ramadan, U., Yla-Herttuala, S., Petrova, T.V., and Alitalo, K. 2007. Therapeutic differentiation and maturation of lymphatic vessels after lymph node dissection and transplantation. Nat. Med. 13: 1458-1466.

Tammela, T., Saaristo, A., Lohela, M., Morisada, T., Tornberg, J., Norrmen, C., Oike, Y., Pajusola, K., Thurston, G., Suda, T. et al. 2005. Angiopoietin-1 promotes lymphatic sprouting and hyperplasia. Blood 105: 4642-4648.

Page 123: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

122

Taniguchi, K., Kohno, R., Ayada, T., Kato, R., Ichiyama, K., Morisada, T., Oike, Y., Yonemitsu, Y., Maehara, Y., and Yoshimura, A. 2007. Spreds are essential for embryonic lymphangiogenesis by regulating vascular endothelial growth factor receptor 3 signaling. Mol. Cell. Biol. 27: 4541-4550.

Thielitz, A. and Gollnick, H. 2008. Topical retinoids in acne vulgaris: update on efficacy and safety. Am. J. Clin. Dermatol. 9: 369-381.

Tobler, N.E. and Detmar, M. 2006. Tumor and lymph node lymphangiogenesis--impact on cancer metastasis. J. Leukoc. Biol. 80: 691-696.

Tomarev, S.I., Sundin, O., Banerjee-Basu, S., Duncan, M.K., Yang, J.M., and Piatigorsky, J. 1996. Chicken homeobox gene Prox 1 related to Drosophila prospero is expressed in the developing lens and retina. Dev. Dyn. 206: 354-367.

Trzewik, J., Mallipattu, S.K., Artmann, G.M., Delano, F.A., and Schmid-Schonbein, G.W. 2001. Evidence for a second valve system in lymphatics: endothelial microvalves. FASEB J. 15: 1711-1717.

Tsimboukis, S., Merikas, I., Karapanagiotou, E.M., Saif, M.W., and Syrigos, K.N. 2009. Erlotinib-induced skin rash in patients with non-small-cell lung cancer: pathogenesis, clinical significance, and management. Clin. Lung Cancer 10: 106-111.

Vegetti, M. 1995. The epistemology of Erasistrate and Hellenistic technology. Clio Med. 28: 461-472.

Veikkola, T., Jussila, L., Makinen, T., Karpanen, T., Jeltsch, M., Petrova, T.V., Kubo, H., Thurston, G., McDonald, D.M., Achen, M.G. et al. 2001. Signalling via vascular endothelial growth factor receptor-3 is sufficient for lymphangiogenesis in transgenic mice. EMBO J. 20: 1223-1231.

Vittet, D., Prandini, M.H., Berthier, R., Schweitzer, A., Martin-Sisteron, H., Uzan, G., and Dejana, E. 1996. Embryonic stem cells differentiate in vitro to endothelial cells through successive maturation steps. Blood 88: 3424-3431.

Vlahakis, N.E., Young, B.A., Atakilit, A., and Sheppard, D. 2005. The lymphangiogenic vascular endothelial growth factors VEGF-C and -D are ligands for the integrin alpha9beta1. J. Biol. Chem. 280: 4544-4552.

Von der Weid, P.Y. and Zawieja, D.C. 2004. Lymphatic smooth muscle: the motor unit of lymph drainage. Int. J. Biochem. Cell Biol. 36: 1147-1153.

Von Minckwitz, G., Jonat, W., Fasching, P., du Bois, A., Kleeberg, U., Luck, H.J., Kettner, E., Hilfrich, J., Eiermann, W., Torode, J. et al. 2005. A multicentre phase II study on gefitinib in taxane- and anthracycline-pretreated metastatic breast cancer. Breast Cancer Res. Treat. 89: 165-172.

Wang, J.F., Zhang, X.F., and Groopman, J.E. 2001. Stimulation of beta 1 integrin induces tyrosine phosphorylation of vascular endothelial growth factor receptor-3 and modulates cell migration. J. Biol. Chem. 276: 41950-41957.

Wartenberg, M., Donmez, F., Ling, F.C., Acker, H., Hescheler, J., and Sauer, H. 2001. Tumor-induced angiogenesis studied in confrontation cultures of multicellular tumor spheroids and embryoid bodies grown from pluripotent embryonic stem cells. FASEB J. 15: 995-1005.

Wartenberg, M., Gunther, J., Hescheler, J., and Sauer, H. 1998. The embryoid body as a novel in vitro assay system for antiangiogenic agents. Lab. Invest. 78: 1301-1314.

Wetterwald, A., Hoffstetter, W., Cecchini, M.G., Lanske, B., Wagner, C., Fleisch, H., and Atkinson, M. 1996. Characterization and cloning of the E11 antigen, a marker expressed by rat osteoblasts and osteocytes. Bone 18: 125-132.

White, J., Highland, M., and Clagett-Dame, M. 2000. Abnormal development of the sinuatrial venous valve and posterior hindbrain may contribute to late fetal resorption of vitamin A-deficient rat embryos. Teratology 62: 374-384.

White, J.C., Shankar, V.N., Highland, M., Epstein, M.L., DeLuca, H.F., and Clagett-Dame, M. 1998. Defects in embryonic hindbrain development and fetal resorption resulting from

Page 124: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

123

vitamin A deficiency in the rat are prevented by feeding pharmacological levels of all-trans-retinoic acid. Proc. Natl. Acad. Sci. U. S. A. 95: 13459-13464.

Wicki, A., Lehembre, F., Wick, N., Hantusch, B., Kerjaschki, D., and Christofori, G. 2006. Tumor invasion in the absence of epithelial-mesenchymal transition: podoplanin-mediated remodeling of the actin cytoskeleton. Cancer Cell 9: 261-272.

Wigle, J.T., Chowdhury, K., Gruss, P., and Oliver, G. 1999. Prox1 function is crucial for mouse lens-fibre elongation. Nat. Genet. 21: 318-322.

Wigle, J.T., Harvey, N., Detmar, M., Lagutina, I., Grosveld, G., Gunn, M.D., Jackson, D.G., and Oliver, G. 2002. An essential role for Prox1 in the induction of the lymphatic endothelial cell phenotype. EMBO J. 21: 1505-1513.

Wilting, J., Aref, Y., Huang, R., Tomarev, S.I., Schweigerer, L., Christ, B., Valasek, P., and Papoutsi, M. 2006. Dual origin of avian lymphatics. Dev. Biol. 292: 165-173.

Wilting, J., Tomarev, S.I., Christ, B., and Schweigerer, L. 2003. Lymphangioblasts in embryonic lymphangiogenesis. Lymphat. Res. Biol. 1: 33-40.

Wobus, A.M. and Boheler, K.R. 2005. Embryonic stem cells: prospects for developmental biology and cell therapy. Physiol. Rev. 85: 635-678.

Wong, R.W. 2003. Transgenic and knock-out mice for deciphering the roles of EGFR ligands. Cell. Mol. Life Sci. 60: 113-118.

Wynd, S., Carron, J., Selve, B., Leggat, P.A., Melrose, W., and Durrheim, D.N. 2007. Qualitative analysis of the impact of a lymphatic filariasis elimination programme using mass drug administration on Misima Island, Papua New Guinea. Filaria J. 6: 1.

Yaniv, K., Isogai, S., Castranova, D., Dye, L., Hitomi, J., and Weinstein, B.M. 2006. Live imaging of lymphatic development in the zebrafish. Nat. Med. 12: 711-716.

Yarden, Y. and Sliwkowski, M.X. 2001. Untangling the ErbB signalling network. Nature Rev. 2: 127-137.

Yoon, C.M., Hong, B.S., Moon, H.G., Lim, S., Suh, P.G., Kim, Y.K., Chae, C.B., and Gho, Y.S. 2008. Sphingosine-1-phosphate promotes lymphangiogenesis by stimulating S1P1/Gi/PLC/Ca2+ signaling pathways. Blood 112: 1129-1138.

Young, P.E., Baumhueter, S., and Lasky, L.A. 1995. The sialomucin CD34 is expressed on hematopoietic cells and blood vessels during murine development. Blood 85: 96-105.

Yuan, L., Moyon, D., Pardanaud, L., Breant, C., Karkkainen, M.J., Alitalo, K., and Eichmann, A. 2002. Abnormal lymphatic vessel development in neuropilin 2 mutant mice. Development 129: 4797-4806.

Zhang, S.C., Wernig, M., Duncan, I.D., Brustle, O., and Thomson, J.A. 2001. In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat. Biotechnol. 19: 1129-1133.

Zhao, X., Liu, J., and Ahmad, I. 2002. Differentiation of embryonic stem cells into retinal neurons. Biochem. Biophys. Res. Commun. 297: 177-184.

Zlotnik, A. 2004. Chemokines in neoplastic progression. Semin. Cancer Biol. 14: 181-185.

Page 125: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

124

8 CURRICULUM VITAE

Daniela Marino Neunbrunnenstrasse 215, 8046 Zurich, Switzerland Telephone: +41 44 633 7326 (Office); +41 76 455 31 54 (Mobile) Email: [email protected]; [email protected] Personal Data Date of birth: November 17, 1981 Place of birth: Agrigento, Italy Nationality: Italian Marital status: Unmarried

Education 09/2005-present PhD studies under supervision of Prof. Michael Detmar Swiss Federal Institute of Technology (ETH) Zurich, Switzerland 07/2005 Master in Biotechnology, Functional Genomics and Bioinformatics University of Milan, Italy Thesis: “Study and analysis of learning, differentiation and proliferation of human neural stem cells” Stem Cell Research Institute (SCRI), San Raffaele Hospital, Milan, Italy 07/2003 Bachelor in Biotechnology University of Milan, Italy Thesis: “An hybrid biologic-electronic neural network experiment and analysis” Stem Cell Research Institute (SCRI), San Raffaele Hospital, Milan, Italy 07/2000 High school graduation Liceo Scientifico Statale, A. Sciascia, Canicattì, Agrigento, Italy Work Experience 09/2005-present PhD thesis project in the group of Prof. Michael Detmar Swiss Federal Institute of Technology (ETH) Zurich, Switzerland Thesis: “Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

embryonic stem cell-derived embryoid body assay”.

Page 126: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

125

08/2004-09/2004 Teaching of molecular and cell biology, genetics, evolution and anatomy for high school students

Study center, G. Toniolo, Canicattì, Agrigento, Italy Further Education and Workshops BD-Swiss Tissue Culture Society workshop, “Cell culture models in Pharma Research”. Basel,

Switzerland. November, 2008. “Genetics, breeding and monitoring of mutant mice”. University of Zurich, Switzerland. November,

2007 Laboratory Animal Science, theoretical and practical training in laboratory animal handling. University of Zurich, Switzerland. September, 2006. PALM MicroBeam workshop. Istituto Oncologico della Svizzera Italiana (IOSI), Bellinzona,

Switzerland. February, 2006. “In vitro culture, Embryos, ES –cells, Primary Cells”. University of Zurich, Switzerland. October, 2005. Other Professional Activities 2006-2009 Head of the social committee of the Pharmaceutical Scientists’ Association (PSA) at the

Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland 2006-2008 Teaching assistant, yearly one-week practical course in medicinal chemistry for Bachelor

students in Pharmaceutical Sciences at the Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland

2007 Supervisor of semester- and diploma-projects of students in Pharmaceutical Sciences Publications and Patent R.M.R. Pizzi, D. Rossetti, G. Cino, D. Marino, A.L.Vescovi and W. Baer (2009). A cultured human

neural network operates a robotic actuator. Biosystems 95, 137-144. D. Marino, V. Dabouras, A.W. Braendli and M. Detmar. A role for all-trans-retinoic acid in the early

steps of lymphatic vascular development. Submitted D. Marino, Y. Angehrn, S. Riccardi, N. Baenziger-Tobler, M.R. Pittelkow and M. Detmar. Activation of

the epidermal growth factor receptor promotes lymphangiogenesis in vitro and in vivo. Manuscript in preparation.

D. M. Leslie-Pedrioli, V. Dabouras, T. Karpanen, G. Jurisic, J.W. Shin, D. Marino, R. E. Kalin, S.

Leidel, P. Cinelli, S. Schulte-Merker, A. W. Braendli and M. Detmar. miR-31 inhibits lymphatic lineage-specific differentiation in vitro and lymphatic vessel development in vivo. Submitted.

L. Cueni, L. Chen, D. Marino, R. Hunggenberger, A. Alitalo and M. Detmar. Podoplanin-Fc reduces

lymphatic vessel formation in vitro and in vivo and causes disseminated intravascular coagulation when transgenically expressed in the skin. In revision.

Page 127: Identification of molecular mechanisms controlling lymphatic vessel formation by use of the

126

Pending patent: Multiwell culture plate for three-dimensional cultures. Priority date: 20.02.08. http://www.wipo.int, patent database, code: WO/2009/103416

Conference Presentations

With ORAL presentation: SSCN - Swiss Stem Cell Network annual meeting

D. Marino, Y. Angehrn, M. Detmar. “Lymphatic endothelial cell differentiation of mouse embryonic stem cells” Organizer: Prof. Lukas Sommer; University of Zurich, Switzerland. December 14, 2007.

With POSTER presentation: SSCN- Swiss Stem Cell Network annual meeting

D. Marino, V. Dabouras, A.W. Braendli and M. Detmar. “All-trans-RA is involved in the early processes of lymphatic vascular system development”. Organizers: Brigitte Galliot, Marcos Gonzalez Gaïtan; University of Geneva, Switzerland. February 6, 2009.

ESDR – European Society for Dermatological Research D. Marino, R. Liersch, M. Detmar. “Induction of lymphatic endothelial cell differentiation in mouse embryonic stem cells” Organizer: Reinhard Dummer; University of Zurich, Switzerland. September 5–8, 2007.

KEYSTONE Symposium - Stem Cells and Cancer D. Marino, R. Liersch, M. Detmar. “Induction of lymphatic endothelial cell differentiation in mouse embryonic stem cells” Organizers: Irving L. Weissman and Michael F. Clarke; Keystone Resort, Keystone, Colorado, USA. March 2-7, 2007.

EuroStemCell International Conference - Advances in Stem Cell Research D. Marino, R. Liersch, M. Bruderer, C. Bader, M. Detmar. “Lymphatic endothelial cell differentiation of mouse embryonic stem cells” Organisers: Yann Barrandon, Austin Smith, Elena Cattaneo; Ecole Polytechnique Fédérale de Lausanne, Switzerland. September 8-10, 2006.

ESH - European School of Haematology, Euroconference on Angiogenesis D. Marino, R. Liersch, M. Bruderer, C. Bader, M. Detmar. “Lymphatic endothelial cell differentiation of mouse embryonic stem cells” Organizers: K. Alitalo, Ch. Betsholtz, A. Bikfalvi, C. Rüegg, G. Tobelem; Mandelieu/Cannes, France. May 13-16, 2006.

Outside Interestes and Activities Sports (aqua-fitness, yoga, volleyball, kickboxing, snowboarding), travelling, cooking, reading, singing,

new technologies.