the role of the angiopoietins in vascular morphogenesis

13
ORIGINAL PAPER The role of the Angiopoietins in vascular morphogenesis Markus Thomas Hellmut G. Augustin Received: 3 February 2009 / Accepted: 24 April 2009 / Published online: 16 May 2009 Ó Springer Science+Business Media B.V. 2009 Abstracts The Angiopoietin/Tie system acts as a vascu- lar specific ligand/receptor system to control endothelial cell survival and vascular maturation. The Angiopoietin family includes four ligands (Angiopoietin-1, Angiopoie- tin-2 and Angiopoietin-3/4) and two corresponding tyro- sine kinase receptors (Tie1 and Tie2). Ang-1 and Ang-2 are specific ligands of Tie2 binding the receptor with similar affinity. Tie2 activation promotes vessel assembly and maturation by mediating survival signals for endothelial cells and regulating the recruitment of mural cells. Ang-1 acts in a paracrine agonistic manner inducing Tie2 phos- phorylation and subsequent vessel stabilization. In contrast, Ang-2 is produced by endothelial cells and acts as an autocrine antagonist of Ang-1-mediated Tie2 activation. Ang-2 thereby primes the vascular endothelium to exoge- nous cytokines and induces vascular destabilization at higher concentrations. Ang-2 is strongly expressed in the vasculature of many tumors and it has been suggested that Ang-2 may act synergistically with other cytokines such as vascular endothelial growth factor to promote tumor- associated angiogenesis and tumor progression. The better mechanistic understanding of the Ang/Tie system is grad- ually paving the way toward the rationale exploitation of this vascular signaling system as a therapeutic target for neoplastic and non-neoplastic diseases. Keywords Angiogenesis Endothelial cell Angiopoietin Tie Structure and expression of Angiopoietin ligands and Tie receptors Structure and expression of Angiopoietin-1 and Angiopoietin-2 The Angiopoietins have been identified in the mid 1990s as a family of growth factors that are essential for blood vessel formation (Fig. 1). There are four Angiopoietins known, Angiopoietin-1 (Ang-1), Angiopoietin-2 (Ang-2), Angiopoietin-3 (Ang-3), and Angiopoietin-4 (Ang-4). The best characterized Angiopoietins are Ang-1 and Ang-2. Ang-3 and Ang-4 are orthologs found in mouse and human, respectively. The Angiopoietins are all ligands for the Tie2 receptor [15]. Structurally, the Angiopoietins are com- posed of two domains. There is a N-terminal coiled-coil domain which is responsible for ligand homo-oligomeri- zation of the ligands. Electron microscopy experiments have demonstrated that Ang-1 and Ang-2 can form heter- ogeneous multimers with trimers, tetramers and pentamers [6]. Furthermore, oligomerization is necessary for receptor activation but not for receptor binding. This is mediated by the fibrinogen-like domain which is located in the C-ter- minus [1, 7]. The Angiopoietins are secreted glycoproteins with a dimeric molecular weight of approximately 75 kDa. Ang-1 M. Thomas H. G. Augustin (&) Joint Research Division Vascular Biology, Medical Faculty Mannheim (CBTM), University of Heidelberg, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany e-mail: [email protected] Present Address: M. Thomas Roche Diagnostics GmbH, Pharma Research Penzberg, Penzberg, Germany M. Thomas H. G. Augustin German Cancer Research Center Heidelberg (DKFZ-ZMBH Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany 123 Angiogenesis (2009) 12:125–137 DOI 10.1007/s10456-009-9147-3

Upload: markus-thomas

Post on 15-Jul-2016

232 views

Category:

Documents


15 download

TRANSCRIPT

Page 1: The role of the Angiopoietins in vascular morphogenesis

ORIGINAL PAPER

The role of the Angiopoietins in vascular morphogenesis

Markus Thomas Æ Hellmut G. Augustin

Received: 3 February 2009 / Accepted: 24 April 2009 / Published online: 16 May 2009

� Springer Science+Business Media B.V. 2009

Abstracts The Angiopoietin/Tie system acts as a vascu-

lar specific ligand/receptor system to control endothelial

cell survival and vascular maturation. The Angiopoietin

family includes four ligands (Angiopoietin-1, Angiopoie-

tin-2 and Angiopoietin-3/4) and two corresponding tyro-

sine kinase receptors (Tie1 and Tie2). Ang-1 and Ang-2 are

specific ligands of Tie2 binding the receptor with similar

affinity. Tie2 activation promotes vessel assembly and

maturation by mediating survival signals for endothelial

cells and regulating the recruitment of mural cells. Ang-1

acts in a paracrine agonistic manner inducing Tie2 phos-

phorylation and subsequent vessel stabilization. In contrast,

Ang-2 is produced by endothelial cells and acts as an

autocrine antagonist of Ang-1-mediated Tie2 activation.

Ang-2 thereby primes the vascular endothelium to exoge-

nous cytokines and induces vascular destabilization at

higher concentrations. Ang-2 is strongly expressed in the

vasculature of many tumors and it has been suggested that

Ang-2 may act synergistically with other cytokines such as

vascular endothelial growth factor to promote tumor-

associated angiogenesis and tumor progression. The better

mechanistic understanding of the Ang/Tie system is grad-

ually paving the way toward the rationale exploitation of

this vascular signaling system as a therapeutic target for

neoplastic and non-neoplastic diseases.

Keywords Angiogenesis � Endothelial cell �Angiopoietin � Tie

Structure and expression of Angiopoietin ligands

and Tie receptors

Structure and expression of Angiopoietin-1

and Angiopoietin-2

The Angiopoietins have been identified in the mid 1990s as

a family of growth factors that are essential for blood

vessel formation (Fig. 1). There are four Angiopoietins

known, Angiopoietin-1 (Ang-1), Angiopoietin-2 (Ang-2),

Angiopoietin-3 (Ang-3), and Angiopoietin-4 (Ang-4). The

best characterized Angiopoietins are Ang-1 and Ang-2.

Ang-3 and Ang-4 are orthologs found in mouse and human,

respectively. The Angiopoietins are all ligands for the Tie2

receptor [1–5]. Structurally, the Angiopoietins are com-

posed of two domains. There is a N-terminal coiled-coil

domain which is responsible for ligand homo-oligomeri-

zation of the ligands. Electron microscopy experiments

have demonstrated that Ang-1 and Ang-2 can form heter-

ogeneous multimers with trimers, tetramers and pentamers

[6]. Furthermore, oligomerization is necessary for receptor

activation but not for receptor binding. This is mediated by

the fibrinogen-like domain which is located in the C-ter-

minus [1, 7].

The Angiopoietins are secreted glycoproteins with a

dimeric molecular weight of approximately 75 kDa. Ang-1

M. Thomas � H. G. Augustin (&)

Joint Research Division Vascular Biology, Medical Faculty

Mannheim (CBTM), University of Heidelberg, Im Neuenheimer

Feld 280, 69120 Heidelberg, Germany

e-mail: [email protected]

Present Address:M. Thomas

Roche Diagnostics GmbH, Pharma Research Penzberg,

Penzberg, Germany

M. Thomas � H. G. Augustin

German Cancer Research Center Heidelberg (DKFZ-ZMBH

Alliance), Im Neuenheimer Feld 280, 69120 Heidelberg,

Germany

123

Angiogenesis (2009) 12:125–137

DOI 10.1007/s10456-009-9147-3

Page 2: The role of the Angiopoietins in vascular morphogenesis

has 498 aa and is located on chromosome 8q22. Ang-2 has

496 aa and is located on chromosome 8q23. Both molecules

show sequence homology of about 60% [1, 2]. Ang-1 is

expressed by smooth muscle cells and other perivascular

cells. Like Ang-2, it binds Tie2 with an affinity of about 3 nM

[2] at the IgG-like domain and the EGF-like domain of Tie2

[8]. Ang-1 is produced as four different splice variants. The

splice variants with 1.5 kb (full length Ang-1) and 1.3 kb

bind the receptor and induce its autophosphorylation. The

proteins coded by the 0.9 kb and 0.7 kb also bind Tie2, but do

not induce autophosphorylation [9]. A novel Ang-2 splice

variant, Ang-2B, with a truncated amino-terminal domain

has been detected in chicken [10]. An additional splice var-

iant (Ang-2(443)) has been identified which lacks parts of the

coiled-coil domain and cannot stimulate Tie2 phosphoryla-

tion [11]. Ang-1 acts as an agonist of the Tie2 receptor,

whereas Ang-2 is the antagonist [2]. However, Ang-2 has

also been reported to context-dependently induce receptor

phosphorylation. The molecular basis for agonistic versus

antagonistic functions of Ang-2 have not been unraveled.

Cell type specific effects, the degree of endothelial conflu-

ence, the duration of Ang-2 stimulation, concentration-

dependent effects, as well as the presence of co-receptors

such as Tie1 have all been implicated in controlling agonistic

versus antagonistic functions of Ang-2 [12–14].

Ang-2 is almost exclusively expressed by endothelial

cells where it is stored in Weibel-Palade bodies (WPB)

[15]. Following cytokine activation of the endothelium

(e.g., by Histamine or Thrombin), Ang-2 is rapidly released

from WPB [15]. It acts in an autocrine manner on the Tie2

receptor by binding as homodimers or multimers [7].

Recent studies have shown that endogenous Ang-2 may act

through an internal autocrine loop mechanism. This con-

cept is based on cellular experiments showing that

endogenously released Ang-2 cannot be inhibited by

exogenous soluble Tie2 receptor [16].

Ang-2 levels are upregulated by hypoxia [17–20]. Under

physiological conditions, Ang-2 is expressed in regions of

vascular remodeling, for example during vascularization of

the retina or during vessel regression in the cyclic ovarian

corpus luteum [2, 21]. Ang-2 expression is also upregulated

under pathological condition, e.g., in the endothelium of

tumors [22–24] and in the tumor cells themselves [25–27].

Moreover, retinal neurons [28] and Muller cells [29] are a

source of Ang-2.

In contrast to Ang-2, Ang-1 is primarily expressed by

mesenchymal cells and acts in a paracrine manner on the

endothelium. It is abundantly expressed by the myocar-

dium during early development and by perivascular cells

later during development and in adult tissues [2, 30, 31].

Ang-1 is also expressed by tumor cells [22, 32] and neu-

ronal cells of the brain [32].

Expression and structure of Tie1 and Tie2

Tie1 and Tie2 are endothelial cell-specific receptors with

similar molecular weight of approximately 135 and

150 kDa, respectively. Originally identified as orphan

receptors in the early nineteen nineties (Fig. 1), they are

expressed by vascular and lymphatic endothelial cells.

Both receptors are structurally similar in the cytoplasmic

region (76% sequence identity), but show only 33% simi-

larity in the extracellular part [33]. Tie1 and Tie2 are

tyrosine kinases with Ig-like and EGF-like homology

1992 1993 1994 1995 1996 1997 1998 1999 2000 2001 2002 2003 2004 2005 2006 2007 2008

Ang-1 actsthrough Akt

Ang-2 hasagonistic function

Generation ofTie2-Cre mice

Role of Ang-2 as acontextual remodelling

molecule

Role of Ang-2 role indiabetic retinopathy

Ang-1 restores vesselarchitecture in absence

of mural cells

Ang-2 nullmice

Ang-2 stored inWP bodies

Tie2-positivemonocytes

Low affinity Ang-1 /Tie1 binding

Ang-1 stimulateslymphangiogenesis

Destabilizing role ofautocrine Ang-2

Tie1 cleavage byshear stress

Ang-2 survival factorin stressed EC

Ang-2 role in hyperoxia-induced lung injury

Ang-1 interactionwith mDia

Ang-2 in macrophagecell death switch

Contextual Tie2signalling

Discoveryof Tie1

Constitutive pTie2in quiescent EC

Role of Tie2 inhematopoiesis

Tie2 associateswith Dok-R

Discoveryof Tie2

Knockout ofTie1 & Tie2

No role of Tie1 inhematopoiesis

Identificationof Ang-2

Identificationof Ang-1

Contextuality modelof Ang-2 function

Ang-1-inducedvascularization

Cooperativity betweenAngs and VEGF

Ang-1 vesselsealing function

Role of Ang-2 invessel cooption

Role of Tie-2 in AVmalformations

Ang-1 as an anti-apotopic molecule

Ang-2 primarilyproduced by EC

Ang-1 and Ang-2molecular structure

Ang-1-inducedEC sprouting

Cytokine regulation ofAng-2 expression

Identification ofAng-3/4

Rec

epto

rsLi

gand

s

Ang-1 reducesinflammatory markers

Ang-1 protects againstplasma leakage

Ang-1 controls thestem cell niche

Ang1-FOXO1 axisestablished

COMP-Ang-1developed

Ang-2 facilitates ECresponsiveness to

inflammatory stimuli

Fig. 1 A possible mechanism for the initiation of angiogenesis

receptors in the early 1990s, the angiopoietin ligands were identified

few years later. Major milestones include the genetic manipulation of

receptors and ligands in loss-of-function and gain-of-function

experiments as well as the unraveling of relevant signaling pathways,

cellular readouts of receptor activation, and adult manipulatory

experiments in neoplastic and non-neoplastic settings

126 Angiogenesis (2009) 12:125–137

123

Page 3: The role of the Angiopoietins in vascular morphogenesis

domains. The extracellular domain consists of three

immunoglobulin (Ig)-like domains that are flanked by three

epidermal growth factor (EGF)-like cysteine repeats fol-

lowed by three fibronectin type III domains (Fig. 2a). The

smaller intracellular domains of both receptors consist of a

split kinase domain which can bind different molecules

after autophosphorylation. Crystal structures analyses

showed that the ligands Ang-1 and Ang-2 bind with almost

similar affinity to the same site of the Tie2 receptor [8].

They bind to the second Ig-like loop flanked by the first Ig-

like loop and the EGF-like repeats [8, 34].

During early development, Tie1 can be detected from

E8.5 in differentiating angioblasts of the head mesen-

chyme, in the splanchnopleura and in the dorsal aorta but

also in migrating endothelial cells of the developing heart

[35]. Tie1 is almost exclusively expressed by endothelial

cells. Full length Tie1 is thought to heterodimerize with

Tie2 [36]. Other studies have shown that the ectodomain of

Tie1 is proteolytically cleaved following VEGF stimula-

tion. The membrane-anchored cleaved form, containing the

cytoplasmic domain, may interact with Tie2 and is sup-

posed to be involved in Tie2 signaling [36–38]. Receptor

shedding also occurs after stimulation with the phorbol

ester PMA, stimulation with tumor necrosis factor alpha

(TNF-a), and by shear stress [39–41]. Tie1 is still largely

considered as an orphan receptor. Yet, recent work sug-

gests that COMP-Ang-1, a designed pentameric form of

Ang-1, can bind to Tie1 under certain conditions [42].

The second Angiopoietin receptor, Tie2, is expressed by

endothelial cells as well as by hematopoietic cells, endo-

thelial precursor cells [43, 44] and tumor cells (e.g., Kaposi

sarcoma cells [45] and melanoma cells [46]). A Tie2-

positive subpopulation of monocytes is associated with the

angiogenic activity of recruited tumor-associated macro-

phages [47]. Endothelial cells in larger vessels express Tie2

more abundantly when compared with smaller vessels [33,

43]. Tie2 expression is upregulated during tumor angio-

genesis [48–50]. The receptor dimerizes by ligand binding.

Following binding of the activating ligand Ang-1, Tie2 is

autophosphorylated and intracellular signaling pathways

are activated (Fig. 3a).

Physiological roles of Tie receptors and Angiopoietin

ligands during development and in the adult

Tie receptors

Tie2-deficient mouse embryos die at E10.5 due to vessel

remodeling defects in the plexus of the yolk sac, of the

brain and severe heart defects. The mice show 30 and 75%

less endothelial cells at E8.5 and E9.5, respectively. Ves-

sels are only poorly organized, have fewer branches and

have reduced pericyte coverage [30, 44, 51, 52]. Tie2 also

exerts critical roles during hematopoiesis [53]. Loss of Tie2

function leads to endothelial cell apoptosis which in turn

results in hemorrhage [54]. These results suggest that the

Ang-Tie system plays a key role during vessel remodeling,

maturation and stabilization of the cardiovascular system.

Injection of soluble Tie2 (sTie2-Fc) was shown to

inhibit ischemia-induced retinal neovascularization in a

mouse model [55]. This soluble form is also present under

physiological conditions in the serum resulting from Tie2

cleavage which has been shown in cellular experiments to

Ig-like

EGF-like

Ig-like

FN III

cell membrane

TK I

TK II

extracellular

intracellular

Tie1/Tie2

A B

Tie1 Tie2

RKTY - 1101VNTTLY - 1107EKFTY - 1112AGI

RKA

1113 - YVNMSL

1119 - FENFT

1124 - YAGI

kinase domain

Grb2

p85

Dok-R

Shp-2

Fig. 2 Schematic overview of

the Tie receptors. a The

extracellular domain of Tie1

and Tie2 consists of three

immunoglobulin (Ig)-like

domains, one EGF-like domain

and three fibrinogen-like

domains. The intracellular part

contains the split kinase

domain. b Tie2 has three

phosphotyrosine residues (1101,

1107, 1112), whereas, Tie1 has

only two (1113 and 1124). The

equivalent to Tie2 pTyr1107 is

missing. Grb2 and p85 both

bind to pTyr1101, Dok-R to

pTyr1107 and SHP2 to

pTyr1112 of Tie2

Angiogenesis (2009) 12:125–137 127

123

Page 4: The role of the Angiopoietins in vascular morphogenesis

occur after PMA stimulation [56]. Circulating concentra-

tions of soluble Tie2 are increased in several vasculopa-

thies, including coronary artery disease [57].

Abnormal vessel structures are not only caused by Tie2-

deficiency. A constitutively active Tie2 mutant has been

identified in patients with venous malformations [58]. This

leads to enlarged veins with pronounced proliferation of

endothelial cells. The endothelial cells are surrounded by

several layers of smooth muscle cells. The range is

between areas with normal coverage and areas completely

devoid of SMC.

Mice lacking the Tie1 gene die between E13.5 and P1

due to a loss of structural integrity of vascular endothelial

cells, resulting in severe edema and hemorrhage [44].

Developmental angiogenesis is not perturbed. Unlike Tie2-

deficient mice, hematopoiesis occurs normally in Tie1-

deficient mice [59]. The genetic experiments suggest that

Tie1 plays important roles during endothelial cell differ-

entiation and in the regulation of vessel integrity.

Double-knockout mice for Tie1 and Tie2 have been

created to shed further light into the signaling pathways of

both receptors during vascular development. These mice

die like Tie2-deficient mice around E10.5 not only due to

cardiovascular defects but also as a consequence of severe

defects in the vascular system. Vasculogenesis proceeds

normally in these mice. The authors concluded from their

results that Tie1 and Tie2 are essential for maintaining the

integrity of mature vessels but that they are dispensable for

early angiogenic sprouting [60].

Angiopoietin ligands

Angiopoietin-1 deficiency results in lethality at E11–E12.5

[30]. The phenotype of Ang-1-deficient mice is similar to

the phenotype of Tie2-deficient mice but not as severe.

These mice have growth-retarded hearts with a less com-

plex ventricular endocardium. The endocardium is col-

lapsed and appears retracted from the myocardial wall. The

endothelial lining in the atria is collapsed and the trabec-

ulae are absent. Ang-1 deficiency also causes severe vas-

cular defects [30]. The mice show a much simpler and

immature primary capillary plexus. The distinction

between larger and smaller vessels is much less pro-

nounced. Periendothelial cells are scarce in Ang-1-deficient

embryos and not associated with endothelial cells but

appear separated from rounded endothelial cells.

Myocardial overexpression of Ang-1 under the control of

the tetracycline promoter shed further light in the importance

of Ang-1 during heart development. Most of these mice

(90%) die between E12.5 and E15.5 as a result of cardiac

hemorrhage. The myocardial walls of both atria and the

ventricles are thinned and the density of trabeculae is dra-

matically reduced. The mice show hemorrhages around the

heart and the atria are enlarged. The outflow tract is collapsed

and mice lack an intact endocardium and coronary arteries.

Ten percent of the mice survive with cardiac hypertrophy

and a dilation of the right atrium [61]. These studies showed

that Ang-1 overexpression dramatically affects early devel-

opment of the mice. Yet, overexpression in the adult has little

effect on vessel structure and heart development.

Transgenic mice overexpressing Ang-1 under the con-

trol of the keratin 14 (K14) promoter are viable and gen-

erally healthy [62]. Newborn mice show larger vessels in

the skin. Additionally, the skin of older mice is more

reddish than those of normal mice. Transmission electron

microscopic analysis confirmed that these mice have nor-

mal cell–cell contacts between endothelial cells and

between endothelial and perivascular cells. The inter-

Fig. 3 Schematic representation of Angiopoietin signaling in regu-

lating the quiescent and the activated phenotype of the endothelium. aAng-1 is produced in non-endothelial cells and binds to Tie2 inducing

Tie2 autophosphorylation. In a next step, PI3-K and Akt are activated

which in turn promotes survival or anti-apoptotic signals through

proteins like, Survivin, Caspase-9, eNOS and Bad. Inactivated FAK

in the cell further supports survival of endothelial cells through Akt.

On the other hand, Rho GTPases are activated by Ang-1 which

reduces endothelial cell permeability by sequestering Src through

mDia. Thereby, VEGF-R2-mediated Src phosphorylation and sub-

sequent VE-cadherin internalization is inhibited. VE-PTP interacts

with Tie2 in the presence, but not in the absence of cell–cell contacts.

VE-PTP inhibition in endothelial cells is associated with increased

permeability. Furthermore, several proteins like Dok-R or Grb14

associate with phosphorylated Tie2 and thereby inhibit endothelial

cell proliferation. Ang-1/Tie2 signaling is required for vessel

stabilization. Ang-2 acts as an antagonistic regulator on endothelial

cells and thereby leads to vessel destabilization and pericyte dropout.

The exact molecular mechanisms of how this process is regulated are

not known. Potential molecules that are involved in this process are

mentioned in the scheme. FOXO transcription factors are also

involved in Ang/Tie signaling by regulating protein synthesis. Their

phosphorylation leads to an inactive form which promotes endothelial

cell survival, quiescence and vascular stabilization, whereas, the

activated form supports vascular destabilization and apoptosis. b Tie2

activation under certain conditions results in cell migration, inflam-

mation and vascular leakage. Cell migration is mediated by the

activation of FAK by PI3-K, adaptor proteins of Dok-R, e.g., Nck and

PAK and by SHP-2, which is thought to dephosphorylate autophos-

phorylation sites of Tie2. Translocation of Tie2 to cell-matrix

attachment sites in subconfluent cells promotes endothelial cell

migration through the activation of Dok-R and its adaptor proteins.

The interaction of ABIN-2 with Tie2 is thought to inactivate NFjB

via the IKK complex and thereby induces destabilization and

inflammation. Rho activation is blocked during Ang/Tie-mediated

vascular leakage, which liberates Src from mDia. VEGF promotes

VEGF-R2 activation which in turn activates Src and induces VE-

cadherin internalization. Abbreviations: Ang, Angiopoietin; SMC,

smooth muscle cell; HB-EGF, heparin-binding epidermal growth

factor-like growth factor; PDGF, platelet-derived growth factor; TGF,

transforming growth factor; BMP, bone morphogenetic protein; Dok-

R, docking protein R; MAPK, mitogen-activated protein kinase;

PAK, p21-activated kinase; PI3-K, phosphatidylinositol 30-kinase;

Akt, protein kinase B; FAK, focal adhesion kinase; eNOS, endothelial

nitric oxide synthase; FKHR, forkhead transcription factor; VE-PTP,

vascular endothelial tyrosine phosphatase

c

128 Angiogenesis (2009) 12:125–137

123

Page 5: The role of the Angiopoietins in vascular morphogenesis

endothelial distance is slightly increased but mice show no

plasma leakage or edema. The experiments demonstrated

that the vasculature is largely intact and functional.

Angiopoietin-2 transgenic mice show severe vascular

defects including disruption of vessel integrity [2]. The

endocardial lining is collapsed and detached from the

underlying myocardium. Trabecular folds are completely

absent. The systemic Ang-2 overexpression phenotype is

highly reminiscent of the phenotype of Ang-1- and Tie2-

deficient mice which supported the hypothesis that Ang-1

P

P

p110p85

Grb2 Akt

FAKCas

Paxillinp85

p110

α vβ3

αvβ3

nucleus

Survivin

Caspase-9

eNOS

Bad

FOXO-1

Rho

Src mDiaSrcP

P

VE-PTP

Tie2

VE-cadherin

Ang-1

Ang-2

extracellular matrix

SerotoninHGF

HB-EGFPDGF-B

TGFβBMP pathway

Dok-R

Ras MAPKcascade

Cellproliferation

Vesselstabilization

Pericyte/SMCrecruitment

nucleus

EC

pericyte

P

P

EC

A

B

VEGFR2VEGF

Abin-2

IKK complex

NFκB NF B

inactive active

inflammation, thrombosis

Rho

extracellular matrix

mDia Src-P VEGFR2-P

VE-cadherin-P

Vascular leakage

Dok-RP

PNck Pak

RasGAP

Shp-2

p85

p110 Grb7P

P

P

P

P

FAK

Cell migrationEC

Tie2

Ang-1

Ang-2

κ

Angiogenesis (2009) 12:125–137 129

123

Page 6: The role of the Angiopoietins in vascular morphogenesis

acts in a stimulating, agonistic manner on Tie2, whereas,

Ang-2 exerts antagonistic functions on Ang-1/Tie2 sig-

naling. Endothelial-specific overexpression of Ang-2 in

adult mice confirmed this hypothesis exhibiting a complete

suppression of Ang-1-mediated Tie2 phosphorylation in

addition to arteriogenesis defects [63]. It has also been

reported that injection of Ang-2 has an effect on pericyte

coverage in the mouse retina. Ang-2 has in these experi-

ments been injected intravitreally which resulted in the

dropout of pericytes [64]. Ang-2 has also been reported to

have different effects dependent on the cytokine milieu.

Ang-2 and VEGF act together to induce angiogenesis.

However, Ang-2 induces vessel regression in the absence/

inhibition of VEGF [2, 65–67].

It has recently been observed that the perinatal lethality

of Ang-2-deficient mice is strain-dependent. Essentially all

Ang-2-deficient mice in the 129/J background die postna-

tally within 14 days after birth [31]. Ang-2-deficient mice

in the C57/Bl6 background are viable with only 10%

postnatal lethality [68]. These mice show no vascular

defects but develop a severe chylous ascites after birth

indicating defects in the lymphatic system. Further analy-

ses revealed that large vessels are disorganized forming a

lacy network with poor smooth muscle cell coverage [69].

Small lymphatic vessels in the intestine are disorganized

and irregular [31]. Ang-2-deficient mice show only minor

vascular defects. Hyaloid vessels in the eye’s lens regress

shortly after birth in wild type, but not in Ang-2-deficient

mice. This reflects a role of Ang-2 in vessel remodeling

and vascular regression [31, 70].

The genetic knock-in of Ang-1 into the Ang-2 locus

completely rescues the lymphatic phenotype of Ang-2-

deficient mice, but not the vascular remodeling defects [31]

supporting the hypothesis that Ang-2 is agonistic in lym-

phatic vessels and antagonistic in blood vessels. These

experiments further support the concept that Ang-2 is

dispensable for early development but necessary for vessel

remodeling and during later stages of development.

Signaling through Tie receptors

Ang-1 and Ang-2 both bind Tie2, but only Ang-1 induces

its autophosphorylation and thereby the activation of the

receptor [1]. As antagonistic ligand, Ang-2 does not induce

receptor autophosphorylation but competes with Ang-1 to

act as an inhibitor of Ang-1/Tie2 signaling [2]. Yet, some

studies also identified Ang-2 as an agonist of Tie2 [12, 14]

Ang-1 binding to Tie2 leads to an activation of signaling

pathways inside the cell by recruiting different adaptor

proteins to the receptor. Signaling is related to several

processes including cell survival, migration, inflammation

and permeability.

Endothelial cell survival and maintenance

Ligand binding of Tie2 leads to phosphorylation of the p85

subunit of phosphatidylinositol 3- kinase (PI3 K). PI3 K

activates Akt which in turn phosphorylates and activates

the Forkhead transcription factor FOXO-1 (FKHR-1).

FKHR-1 is a strong inducer of Ang-2 expression and

inhibits Ang-2 liberation [71–75]. Activation of Akt also

stimulates the phosphorylation and thereby the inhibition

of pro-apoptotic proteins, including BAD and procaspase-9

[72, 76]. Additionally, Akt upregulates survivin, a classical

apoptosis inhibitor, and thereby supports cell survival

(Fig. 3a) [77, 78].

Ang-1- and Tie2-deficient mice show severe defects in

the recruitment of pericytes and in their interaction with

endothelial cells [30, 44, 51]. In a rat model of diabetic

retinopathy, Ang-2 expression was found to be strongly

increased leading to the dropout of pericytes [64]. How-

ever, the mechanisms involved in Ang/Tie-mediated SMC

recruitment are poorly understood. One possible molecule

involved in the recruitment of mural cells is the EC-derived

heparin binding EGF-like growth factor (HB-EGF). Its

expression in endothelial cells is upregulated by Ang-1

[79], but only when they are in contact with mural cells.

HB-EGF-mediated receptor (ErbB1 and ErbB2) activation

thereby induces SMC migration. However, there is also

evidence that hepatocyte growth factor may be involved in

Ang-1-mediated SMC recruitment. Stimulation of endo-

thelial cells with Ang-1 induces SMC migration toward

endothelial cells in a co-culture assay. This effect could be

reversed by the addition of a neutralizing anti-HGF anti-

body indicating that Ang-1 is regulating HGF expression

[80].

In addition to HB-EGF and HGF, PDGF-B is also

expressed by endothelial cells and is involved in pericyte

recruitment [81]. PDGF-B signals through its receptor

PDGFRb which is expressed by pericytes. PDGF-B

thereby acts as chemoattractant which promotes the pro-

liferation of SMCs and pericytes during their recruitment to

the endothelium [82]. PDGFRb antibodies completely

block the recruitment of pericytes to the newly formed

vasculature in the retina of newborn mice leading to retinal

edema and hemorrhage [83]. These vessels are poorly

remodeled and leaky. The injection of recombinant Ang-1

almost completely rescued the phenotype caused by the

blocking of PDGFRb antibodies. This suggests coordinated

activities of Ang-1 and PDGF-B. The vascular defects in

Ang-1- and Tie2-deficient mice occur earlier during

development than those of PDGF-B- and PDGFRb-defi-

cient mice indicating additional mechanisms of pericyte

recruitment. Another regulator of SMC differentiation is

TGF-b which is upregulated by Ang-1 following PDGF-B

stimulation. In turn, Ang-1 is downregulated by TGF-b.

130 Angiogenesis (2009) 12:125–137

123

Page 7: The role of the Angiopoietins in vascular morphogenesis

These data suggest that pericytes are recruited by PDGF-B

which induces pericyte proliferation. Ang-1, which is

upregulated by PDGF-B, promotes pericyte migration.

TGF-b is responsible for SMC differentiation and to render

the vasculature in its quiescent state [84–86]. Ang/Tie

signaling has also been implicated in the regulation of

vessel diameter sensing. The vessel diameter is controlled

in a Tie2-dependent manner by autocrine-acting Apelin

and its cognate receptor APJ [87].

Endothelial cell activation and contextual presentation

of Tie receptors

Tie2 activation has been related to endothelial migration

(Fig. 3B). This seems to be dependent on the contextual

presentation of the receptor on the endothelial cell surface

[88, 89]. Tie2 is expressed in a polarized manner in acti-

vated endothelial cells and translocated to the extracellular

matrix where it binds to matrix immobilized Ang-1. The

Akt pathway is blocked, whereas, Dok-R (docking protein

R) is phosphorylated. Activated Dok-R interacts with ras-

GAP, Nck and Crk. All these molecules are involved in cell

migration, proliferation, cytoskeletal reorganization and

the regulation of the ras signaling cascade [90]. In contrast,

Tie2 is translocated to cell–cell junctions in quiescent

endothelial cells where it engages in trans complexes with

other Tie2 molecules of neighboring cells. In this context,

Tie2 interacts with VE-PTP, a molecule which is strongly

associated with barrier function, thereby inhibiting para-

cellular permeability. Additionally, Akt is activated to

induce endothelial cell survival and stability of the endo-

thelium through the phosphorylation of eNOS (Fig. 3a)

[88, 89]. Src is activated during VEGF-mediated angio-

genesis. This leads to the activation of VAV, a guanine-

nucleotide-exchange factor (GEF) for Rac. Rac further

activates VE-cadherin at Ser665. Beta-arrestin-2 is recrui-

ted to VE-cadherin which leads to its internalization in a

clathrin-dependent manner [91, 92]. This progress supports

vascular permeability and migration. Ang-1-mediated Tie2

signaling inhibits this pathway by activation of mDia

through Rho. This leads to an association of Src and mDia.

Src is not longer available for VE-cadherin activation and

internalization [93]. Ang-1 was further shown to inhibit

Thrombin-induced permeability by decreasing PKCzeta

activation [94]. The same group could show that Ang-1

also prevents vascular permeability in vitro and in vivo by

stimulating sphingosine kinase-1 [95]. Other molecules

like Grb2, Grb7, ShcA, the protein tyrosine kinase SHP2

and the previously mentioned p85 subunit interact with

Tie2 via SH2 domains. These molecules seem to be

involved in cell migration, proliferation, differentiation and

apoptosis [96, 97]. Fusion proteins of the recombinant Tie2

kinase domain and glutathione-S-transferase (GST)

showed that Grb2 interacts with Tie2 at pTyr-1101

(Fig. 2B). A mutation of this tyrosine residue to phenyl-

alanine markedly decreased the interaction of Grb2 with

Tie2. SHP2 association with phosphorylated Tie2 remained

unaffected. Conversely, mutation of pTyr-1112 to phenyl-

alanine reduced the association of SHP2 with the phos-

phorylated kinase domain (Fig. 2b) [96, 98]. These

findings indicate that Grb2 and SHP2 associate with

phosphorylated Tie2, thereby supporting intracellular sig-

naling processes, like activation of MAPK. Indeed, Ang-1

can activate MAPK in endothelial cells and in the aortic

ring assay [99–101]. Yet, the inhibition of MAPK had no

effect on Ang-1-mediated endothelial cell survival and

migration [99].

SHP2 is not only involved in signal transduction. It may

also act as a negative regulator of Tie2 phosphorylation.

Mutation of Tie2 at pTyr1112 enhanced autophosphoryla-

tion and downstream signaling [97, 102]. Yeast-two-hybrid

experiments revealed that the p85 subunit of PI3 K also

interacts with the phosphorylated Tie2 kinase domain [97].

p85 binds like Grb2 to pTyr1101 (Fig. 2b). A mutation of

this phosphorylation site reduced the binding of p85 to

Tie2. Dok-R has been shown to bind to activated Tie2 at

pTyr1107 (Fig 1b). A mutation of this phosphorylation site

reduced the binding of Dok-R to Tie2 [102]. Dok-R is

immediately phophorylated at multiple sites after binding

to activated Tie2 to recruit other molecules. This leads to

the activation of several signaling pathways, including

migration. However, this phosphotyrosine is missing in

Tie1 protein, as shown in Fig. 2b.

Ang-1 is also able to activate focal adhesion kinase (FAK)

through Tie2 [103]. This in turn leads to the phosphorylation

of paxillin. The MAP kinase ERK is activated in further steps

[104] which supports migration. In turn, when blocking Tie2

activation, Ang-1 induced migration via ERK is inhibited.

Endothelial cell sprouting is mediated by the secretion of

plasminogen and metalloproteinase following Ang-1 stim-

ulation [103]. All these in vitro activation phenotypes of

Ang-1 are supported by in vivo studies in mice which have

shown that Ang-1 overexpression promotes vessel formation

in the heart of mice [62].

Role of Ang/Tie signaling during pathology

Angiopoietin-1 functions during inflammation

Ang-1 acts as an anti-inflammatory cytokine. It protects

against endotoxic shock-induced by LPS and thereby pre-

vents microvascular leakage [105]. It blocks the expression

and cell surface activity of tissue factor (TF), an initiator of

blood coagulation, which is involved in thrombosis and

inflammation. Ang-1 reduces VEGF stimulated leukocyte

Angiogenesis (2009) 12:125–137 131

123

Page 8: The role of the Angiopoietins in vascular morphogenesis

adhesion to endothelial cells [106]. Cardiac allograft ath-

erosclerosis [107] and radiation induced cell damage [108]

are protected by Ang-1 and by a designed pentameric Ang-

1, named COMP-Ang-1. Furthermore, Tie2 activation

leads to ABIN-2 recruitment which interferes with NF-jB

signaling [109–111]. This prevents endothelial cells from

undergoing apoptosis and the induction of inflammation.

Subsequent signaling is likely mediated by the PI3 K/Akt

pathway because blocking PI3 K results in suppression of

ABIN-2-induced inhibition of cell death [111]. Ang-1 may

also be involved in inflammatory diseases, like rheumatoid

arthritis (RA). Synovial fibroblasts are a key player during

RA and a major source of Ang-1. Ang-1 is also upregulated

during this disease by inflammation promoting cytokines,

including TNF-a [112, 113]. TNF-a but also IL-1b are

capable to induce the expression of the transcription factor

epithelium-specific Ets-like factor (ESE-1) which is also

detectable in the synovium of RA patients [114]. ESE-1 has

been shown to upregulate Ang-1 indicating that this tran-

scription factor regulates the high Ang-1 mRNA levels

during RA [115].

Angiopoietin-2 functions during inflammation

Little is known about the mechanisms of Ang-2 function on

Tie2. Recent studies have shown that Ang-2 supports

RhoA and MLC activation and thereby promotes vascular

leakage and endothelial cell migration [116]. Other studies

have identified Ang-2 as a pro-inflammatory cytokine.

Ang-2-deficient mice cannot elicit an inflammatory

response in thioglycollate-induced or Staphylococcus aur-

eus-induced peritonitis [68]. Ang-2 serum levels are

increased during sepsis. Normal serum levels are in the

range of 1–2 ng/ml. During sepsis, Ang-2 levels may

increase up to 20 fold (30 ng/ml). Elevated circulating

Ang-2 levels have also been associated with mortality.

More than 50% of patients with soluble Ang-2 levels in

excess, i.e. about 20 ng/ml die [116–119]. Furthermore,

Ang-2 expression correlates with neovascularization during

physiological and pathological processes, like arthritis

[120] or psoriasis [121]. In both diseases, Ang-2 expression

is not only associated with vessel remodeling but also with

VEGF expression. Ang-2 and VEGF act together to induce

angiogenesis and the expression of matrix metalloproteas-

es, proteins that degrade the basement membrane [122].

However, Ang-2 induces vessel regression in the absence/

inhibition of VEGF [2, 65, 66]. Ang-2 increases the

expression level of the matrix metalloprotease MMP-2 in

gliomas which is a sign for active angiogenesis [123].

Moreover, an anti-Ang-2 therapy in the cornea of rats was

shown to inhibit VEGF-induced neovascularization [24].

Ang-2 expression is highly upregulated by angiogenesis-

inducing molecules like VEGF, bFGF or TNF-a.

Thrombin, an angiogenesis promoting molecule, but also

hypoxia, is able to induce Ang-2 expression [17, 19, 20,

124–128]. Hegen and co-workers could show that the

activity of the Ang-2 promoter is regulated by the tran-

scription factor Ets-1 [127]. The implication of Ets-1 in

neovascularization has been shown in a mouse model of

proliferative retinopathy [129]. Ets-1 dominant negative

constructs injected in the eye completely blocked this

function. Its expression is further upregulated by VEGF

and shear stress which in turn increase Ang-2 expression

[130, 131]. Ang-2 expression is also regulated by the

transcription factor FOXO1. This family of transcription

factors is involved in the upregulation of proteins during

destabilization and remodeling. Ang-1 negatively interferes

with FKHR-associated gene expression and thereby sup-

presses the production of Ang-2 [132].

Angiopoietin expression in tumors and tumor

associated angiogenesis

Ang-2 is only weakly expressed in endothelial cells under

physiological conditions. However, Ang-2 expression is

dramatically increased during vascular remodeling, e.g.,

during tumor growth [133]. For example, glioblastoma

show increased levels of Ang-2 in their associated endo-

thelium [32]. Here, Ang-2 is highly expressed in necrotic

and hypoxic regions [26]. Vessels in these areas are not

covered by smooth muscle cells. Only small vessels in

glioblastomas express high amounts of Ang-2 but not

larger ones [32]. Overexpression of Ang-2 in a rat glioma

model resulted in aberrant vessels with low SMC cover-

age [134]. Ang-2 is also detectable in significant con-

centrations in the circulation of tumor patients, e.g., in

esophageal squamous cell cancer [135], hepatocellular

carcinoma [136], and lung cancer [137]. The expression

of Ang-2 in melanomas correlates with tumor progression

[46]. Tumor cells have been shown to express Ang-2,

e.g., stomach [122], colon [138], bladder carcinoma [139],

melanoma [46], and non small cell lung cancer (NSCLC)

[140].

In addition to promoting vessel regression as in glio-

blastomas, Ang-2 induces tumor neovascularization in

combination with angiogenic growth factors such as VEGF

or bFGF. Blocking experiments with Ang-2 neutralizing

antibodies or fusion proteins massively decreased tumor

growth [24]. Antibodies against Ang-2 not only inhibited

Ang-2- but also VEGF-induced endothelial cell migration

and proliferation during angiogenesis [141], which dem-

onstrated enhancing functions of Ang-2 during VEGF-

induced angiogenesis. Moreover, Ang-2 aptamers (RNAs

that bind and thereby block proteins) inhibit bFGF-induced

angiogenesis in the rat corneal assay [142]. In addition to

promoting vessel regression and neovascularization, Ang-2

132 Angiogenesis (2009) 12:125–137

123

Page 9: The role of the Angiopoietins in vascular morphogenesis

can also stimulate breast cancer metastasis in a Tie2-

independent pathway by binding directly to integrin a5b1

[143]. However, Ang-2 overexpression in Lewis lung car-

cinoma and TA3 mammary carcinoma cells suppressed

tumor growth. Angiogenesis was found to be disrupted and

apoptosis was enhanced [144].

The role of Ang-1 in tumor-associated angiogenesis

remains controversial. Ang-1 overexpression leads to

reduced tumor growth in several tumor models [145–147].

Pericyte coverage of the tumor vasculature is massively

increased and thereby stabilized [147, 148]. Yet, Ang-1 has

also been shown to promote tumor growth in rat gliomas

[134] and in plasma cell tumors [149]. The downregulation

of Ang-1 in HeLa cells by antisense RNA inhibited tumor

growth and angiogenesis [150]. These findings suggest that

Angiopoietin-1 promoting or inhibiting functions are

dependent on the tumor cell type, the dosage and possibly

on the amount of Ang-2 in the tumors.

Tie receptor independent signaling and non-vascular

Angiopoietin effects

Several studies support the hypothesis that the Angio-

poietins can activate endothelial or tumor cells in a Tie2-

independent manner. It was shown that endothelial cells

can adhere to immobilized Angiopoietins via avb3 and

a5b1 integrin [151]. The direct binding of Ang-2 to a5b1

stimulates breast cancer metastasis through an a5b1

integrin-mediated pathway via Akt [pS473] [143] and

induces glioma cell invasion by stimulating matrix me-

talloprotease-2 expression through avb1 integrin and FAK

[152]. However, Ang-1 further triggers signaling path-

ways of Tie2 and a5b1 through their interaction in

endothelial cell plated on fibronectin, thereby promoting

angiogenesis [153]. Other studies showed that Ang-1

monomers (that do not activate Tie2) promote cardiac and

skeletal myocyte survival and reduce cardiac hypertrophy

through integrins [154, 155]. It has also been hypothe-

sized that Angiopoietins can interact with integrins

expressed by neuronal cells [156]. However, Ang-1 pro-

motes neurite outgrowth from Tie2-positive dorsal root

ganglion cells and activates PI3 K thereby preventing

neuronal apoptosis [157].

Tie2 is also expressed by a subpopulation of hemato-

poietic stem cells and bone marrow osteoblasts. It has been

shown that Ang-1, produced by osteoblasts, mediates the

adhesion of hematopoietic stem cells to osteoblasts in an

integrin-dependent autocrine manner [158]. Therefore, it

has been suggested that constitutive Ang-1/Tie2 signaling

controls the maintenance of the bone marrow stem cell

niche.

References

1. Davis S, Aldrich TH, Jones PF et al (1996) Isolation of angio-

poietin-1, a ligand for the TIE2 receptor, by secretion-trap

expression cloning. Cell 87:1161–1169. doi:10.1016/S0092-

8674(00)81812-7

2. Maisonpierre PC, Suri C, Jones PF et al (1997) Angiopoietin-2,

a natural antagonist for Tie2 that disrupts in vivo angiogenesis.

Science 277:55–60. doi:10.1126/science.277.5322.55

3. Kim I, Kwak HJ, Ahn JE et al (1999) Molecular cloning and

characterization of a novel angiopoietin family protein, angio-

poietin-3. FEBS Lett 443:353–356. doi:10.1016/S0014-5793

(99)00008-3

4. Nishimura M, Miki T, Yashima R et al (1999) Angiopoietin-3, a

novel member of the angiopoietin family. FEBS Lett 448:254–

256. doi:10.1016/S0014-5793(99)00381-6

5. Valenzuela DM, Griffiths JA, Rojas J et al (1999) Angiopoietins

3 and 4: diverging gene counterparts in mice and humans. Proc

Natl Acad Sci USA 96:1904–1909. doi:10.1073/pnas.96.5.1904

6. Kim KT, Choi HH, Steinmetz MO et al (2005) Oligomerization

and multimerization are critical for angiopoietin-1 to bind and

phosphorylate Tie2. J Biol Chem 280:20126–20131. doi:

10.1074/

jbc.M500292200

7. Procopio WN, Pelavin PI, Lee WM et al (1999) Angiopoietin-1

and -2 coiled coil domains mediate distinct homo-oligomeriza-

tion patterns, but fibrinogen-like domains mediate ligand

activity. J Biol Chem 274:30196–30201. doi:10.1074/jbc.274.

42.30196

8. Fiedler U, Krissl T, Koidl S et al (2003) Angiopoietin-1 and

angiopoietin-2 share the same binding domains in the Tie-2

receptor involving the first Ig-like loop and the epidermal

growth factor-like repeats. J Biol Chem 278:1721–1727. doi:

10.1074/jbc.M208550200

9. Huang YQ, Li JJ, Karpatkin S (2000) Identification of a family

of alternatively spliced mRNA species of angiopoietin-1. Blood

95:1993–1999

10. Mezquita J, Mezquita B, Pau M et al (1999) Characterization of

a novel form of angiopoietin-2 (Ang-2B) and expression of

VEGF and angiopoietin-2 during chicken testicular development

and regression. Biochem Biophys Res Commun 260:492–498.

doi:10.1006/bbrc.1999.0934

11. Kim I, Kim JH, Ryu YS et al (2000) Characterization and

expression of a novel alternatively spliced human angiopoietin-

2. J Biol Chem 275:18550–18556. doi:10.1074/jbc.M910084199

12. Kim I, Kim JH, Moon SO et al (2000) Angiopoietin-2 at high

concentration can enhance endothelial cell survival through the

phosphatidylinositol 30-kinase/Akt signal transduction pathway.

Oncogene 19:4549–4552. doi:10.1038/sj.onc.1203800

13. Teichert-Kuliszewska K, Maisonpierre PC, Jones N et al (2001)

Biological action of angiopoietin-2 in a fibrin matrix model of

angiogenesis is associated with activation of Tie2. Cardiovasc

Res 49:659–670. doi:10.1016/S0008-6363(00)00231-5

14. Daly C, Pasnikowski E, Burova E et al (2006) Angiopoietin-2

functions as an autocrine protective factor in stressed endothelial

cells. Proc Natl Acad Sci USA 103:15491–15496. doi:10.1073/

pnas.0607538103

15. Fiedler U, Scharpfenecker M, Koidl S et al (2004) The Tie-2

ligand angiopoietin-2 is stored in and rapidly released upon

stimulation from endothelial cell Weibel-Palade bodies. Blood

103:4150–4156. doi:10.1182/blood-2003-10-3685

16. Scharpfenecker M, Fiedler U, Reiss Y et al (2005) The Tie-2

ligand angiopoietin-2 destabilizes quiescent endothelium

through an internal autocrine loop mechanism. J Cell Sci

118:771–780. doi:10.1242/jcs.01653

Angiogenesis (2009) 12:125–137 133

123

Page 10: The role of the Angiopoietins in vascular morphogenesis

17. Oh H, Takagi H, Suzuma K et al (1999) Hypoxia and vascular

endothelial growth factor selectively up-regulate angiopoietin-2

in bovine microvascular endothelial cells. J Biol Chem

274:15732–15739. doi:10.1074/jbc.274.22.15732

18. Kim I, Kim JH, Ryu YS et al (2000) Tumor necrosis factor-

alpha upregulates angiopoietin-2 in human umbilical vein

endothelial cells. Biochem Biophys Res Commun 269:361–365.

doi:10.1006/bbrc.2000.2296

19. Huang YQ, Li JJ, Hu L et al (2002) Thrombin induces increased

expression and secretion of angiopoietin-2 from human umbil-

ical vein endothelial cells. Blood 99:1646–1650. doi:10.1182/

blood.V99.5.1646

20. Pichiule P, Chavez JC, LaManna JC (2004) Hypoxic regulation

of angiopoietin-2 expression in endothelial cells. J Biol Chem

279:12171–12180. doi:10.1074/jbc.M305146200

21. Goede V, Schmidt T, Kimmina S et al (1998) Analysis of blood

vessel maturation processes during cyclic ovarian angiogenesis.

Lab Invest 78:1385–1394

22. Zagzag D, Hooper A, Friedlander DR et al (1999) In situ

expression of angiopoietins in astrocytomas identifies angio-

poietin-2 as an early marker of tumor angiogenesis. Exp Neurol

159:391–400. doi:10.1006/exnr.1999.7162

23. Zhang L, Yang N, Park JW et al (2003) Tumor-derived vascular

endothelial growth factor up-regulates angiopoietin-2 in host

endothelium and destabilizes host vasculature, supporting

angiogenesis in ovarian cancer. Cancer Res 63:3403–3412

24. Oliner J, Min H, Leal J et al (2004) Suppression of angiogenesis

and tumor growth by selective inhibition of angiopoietin-2.

Cancer Cell 6:507–516. doi:10.1016/j.ccr.2004.09.030

25. Tanaka S, Mori M, Sakamoto Y et al (1999) Biologic signifi-

cance of angiopoietin-2 expression in human hepatocellular

carcinoma. J Clin Invest 103:341–345. doi:10.1172/JCI4891

26. Koga K, Todaka T, Morioka M et al (2001) Expression of an-

giopoietin-2 in human glioma cells and its role for angiogenesis.

Cancer Res 61:6248–6254

27. Torimura T, Ueno T, Kin M et al (2004) Overexpression of

angiopoietin-1 and angiopoietin-2 in hepatocellular carcinoma. J

Hepatol 40:799–807. doi:10.1016/j.jhep.2004.01.027

28. Hackett SF, Ozaki H, Strauss RW et al (2000) Angiopoietin 2

expression in the retina: upregulation during physiologic and

pathologic neovascularization. J Cell Physiol 184:275–284. doi:

10.1002/1097-4652(200009)184:3\275::AID-JCP1[3.0.CO;2-7

29. Yao D, Taguchi T, Matsumura T et al (2007) High glucose

increases angiopoietin-2 transcription in microvascular endo-

thelial cells through methylglyoxal modification of mSin3A. J

Biol Chem 282:31038–31045. doi:10.1074/jbc.M704703200

30. Suri C, Jones PF, Patan S et al (1996) Requisite role of angio-

poietin-1, a ligand for the TIE2 receptor, during embryonic

angiogenesis. Cell 87:1171–1180. doi:10.1016/S0092-8674(00)

81813-9

31. Gale NW, Thurston G, Hackett SF 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. doi:10.1016/S1534-5807(02)00217-4

32. Stratmann A, Risau W, Plate KH (1998) Cell type-specific

expression of angiopoietin-1 and angiopoietin-2 suggests a role

in glioblastoma angiogenesis. Am J Pathol 153:1459–1466

33. Schnurch H, Risau W (1993) Expression of tie-2, a member of a

novel family of receptor tyrosine kinases, in the endothelial cell

lineage. Development 119:957–968

34. Macdonald PR, Progias P, Ciani B et al (2006) Structure of the

extracellular domain of Tie receptor tyrosine kinases and

localization of the angiopoietin-binding epitope. J Biol Chem

281:28408–28414. doi:10.1074/jbc.M605219200

35. Korhonen J, Polvi A, Partanen J et al (1994) The mouse tie

receptor tyrosine kinase gene: expression during embryonic

angiogenesis. Oncogene 9:395–403

36. Marron MB, Hughes DP, Edge MD et al (2000) Evidence for

heterotypic interaction between the receptor tyrosine kinases

TIE-1 and TIE-2. J Biol Chem 275:39741–39746. doi:

10.1074/jbc.M007189200

37. Tsiamis AC, Morris PN, Marron MB et al (2002) Vascular

endothelial growth factor modulates the Tie-2:Tie-1 receptor

complex. Microvasc Res 63:149–158. doi:10.1006/mvre.2001.

2377

38. Marron MB, Singh H, Tahir TA et al (2007) Regulated prote-

olytic processing of Tie1 modulates ligand responsiveness of the

receptor-tyrosine kinase Tie2. J Biol Chem 282:30509–30517.

doi:10.1074/jbc.M702535200

39. Yabkowitz R, Meyer S, Yanagihara D et al (1997) Regulation of

tie receptor expression on human endothelial cells by protein

kinase C-mediated release of soluble tie. Blood 90:706–715

40. Yabkowitz R, Meyer S, Black T et al (1999) Inflammatory

cytokines and vascular endothelial growth factor stimulate the

release of soluble tie receptor from human endothelial cells via

metalloprotease activation. Blood 93:1969–1979

41. Chen-Konak L, Guetta-Shubin Y, Yahav H et al (2003) Tran-

scriptional and post-translation regulation of the Tie1 receptor

by fluid shear stress changes in vascular endothelial cells.

FASEB J 17:2121–2123

42. Saharinen P, Kerkela K, Ekman N et al (2005) Multiple an-

giopoietin recombinant proteins activate the Tie1 receptor

tyrosine kinase and promote its interaction with Tie2. J Cell Biol

169:239–243. doi:10.1083/jcb.200411105

43. Dumont DJ, Yamaguchi TP, Conlon RA et al (1992) Tek, a

novel tyrosine kinase gene located on mouse chromosome 4, is

expressed in endothelial cells and their presumptive precursors.

Oncogene 7:1471–1480

44. Sato TN, Tozawa Y, Deutsch U et al (1995) Distinct roles of the

receptor tyrosine kinases Tie-1 and Tie-2 in blood vessel for-

mation. Nature 376:70–74. doi:10.1038/376070a0

45. Brown LF, Dezube BJ, Tognazzi K et al (2000) Expression of

Tie1, Tie2, and angiopoietins 1, 2, and 4 in Kaposi’s sarcoma

and cutaneous angiosarcoma. Am J Pathol 156:2179–2183

46. Helfrich I, Edler L, Sucker A et al (2009) Angiopoietin-2 levels

are associated with disease progression in metastatic malignant

melanoma. Clin Cancer Res 15:1384–1392. doi:

10.1158/1078-0432.CCR-08-1615

47. De Palma M, Venneri MA, Galli R et al (2005) Tie2 identifies a

hematopoietic lineage of proangiogenic monocytes required for

tumor vessel formation and a mesenchymal population of peri-

cyte progenitors. Cancer Cell 8:211–226. doi:10.1016/j.ccr.

2005.08.002

48. Wong AL, Haroon ZA, Werner S et al (1997) Tie2 expression

and phosphorylation in angiogenic and quiescent adult tissues.

Circ Res 81:567–574

49. Peters KG, Coogan A, Berry D et al (1998) Expression of Tie2/

Tek in breast tumour vasculature provides a new marker for

evaluation of tumour angiogenesis. Br J Cancer 77:51–56

50. Takahama M, Tsutsumi M, Tsujiuchi T et al (1999) Enhanced

expression of Tie2, its ligand angiopoietin-1, vascular endo-

thelial growth factor, and CD31 in human non-small cell lung

carcinomas. Clin Cancer Res 5:2506–2510

51. Dumont DJ, Gradwohl G, Fong GH et al (1994) Dominant-

negative and targeted null mutations in the endothelial receptor

tyrosine kinase, tek, reveal a critical role in vasculogenesis of

the embryo. Genes Dev 8:1897–1909. doi:10.1101/gad.8.

16.1897

134 Angiogenesis (2009) 12:125–137

123

Page 11: The role of the Angiopoietins in vascular morphogenesis

52. Patan S (1998) TIE1 and TIE2 receptor tyrosine kinases inver-

sely regulate embryonic angiogenesis by the mechanism of

intussusceptive microvascular growth. Microvasc Res 56:1–21.

doi:10.1006/mvre.1998.2081

53. Takakura N, Huang XL, Naruse T et al (1998) Critical role of

the TIE2 endothelial cell receptor in the development of defin-

itive hematopoiesis. Immunity 9:677–686. doi:10.1016/S1074-

7613(00)80665-2

54. Jones N, Voskas D, Master Z et al (2001) Rescue of the early

vascular defects in Tek/Tie2 null mice reveals an essential

survival function. EMBO Rep 2:438–445

55. Takagi H, Koyama S, Seike H et al (2003) Potential role of the

angiopoietin/tie2 system in ischemia-induced retinal neovascu-

larization. Invest Ophthalmol Vis Sci 44:393–402. doi:

10.1167/iovs.02-0276

56. Reusch P, Barleon B, Weindel K et al (2001) Identification of a

soluble form of the angiopoietin receptor TIE-2 released from

endothelial cells and present in human blood. Angiogenesis

4:123–131. doi:10.1023/A:1012226627813

57. Chung NA, Makin AJ, Lip GY (2003) Measurement of the soluble

angiopoietin receptor tie-2 in patients with coronary artery dis-

ease: development and application of an immunoassay. Eur J Clin

Invest 33:529–535. doi:10.1046/j.1365-2362.2003.01173.x

58. Vikkula M, Boon LM, Carraway KL 3rd et al (1996) Vascular

dysmorphogenesis caused by an activating mutation in the

receptor tyrosine kinase TIE2. Cell 87:1181–1190. doi:10.1016/

S0092-8674(00)81814-0

59. Rodewald HR, Sato TN (1996) Tie1, a receptor tyrosine kinase

essential for vascular endothelial cell integrity, is not critical for

the development of hematopoietic cells. Oncogene 12:397–404

60. Puri MC, Partanen J, Rossant J et al (1999) Interaction of the

TEK and TIE receptor tyrosine kinases during cardiovascular

development. Development 126:4569–4580

61. Ward NL, Van Slyke P, Sturk C et al (2004) Angiopoietin 1

expression levels in the myocardium direct coronary vessel

development. Dev Dyn 229:500–509. doi:10.1002/dvdy.10479

62. Suri C, McClain J, Thurston G et al (1998) Increased vascu-

larization in mice overexpressing angiopoietin-1. Science

282:468–471. doi:10.1126/science.282.5388.468

63. Reiss Y, Droste J, Heil M, Tribulova S et al (2007) Angio-

poietin-2 impairs revascularization after limb ischemia. Circ Res

101:88–96. doi:10.1161/CIRCRESAHA.106.143594

64. Hammes HP, Lin J, Wagner P et al (2004) Angiopoietin-2

causes pericyte dropout in the normal retina: evidence for

involvement in diabetic retinopathy. Diabetes 53:1104–1110.

doi:10.2337/diabetes.53.4.1104

65. Holash J, Maisonpierre PC, SJ ComptonD et al (1999) Vessel

cooption, regression, and growth in tumors mediated by angio-

poietins and VEGF. Science 284:1994–1998

66. Lobov IB, Brooks PC, Lang RA (2002) Angiopoietin-2 displays

VEGF-dependent modulation of capillary structure and endo-

thelial cell survival in vivo. Proc Natl Acad Sci USA 99:11205–

11210. doi:10.1073/pnas.172161899

67. Korff T, Kimmina S, Martiny-Baron G et al (2001) Blood vessel

maturation in a 3-dimensional spheroidal coculture model:

direct contact with smooth muscle cells regulates endothelial

cell quiescence and abrogates VEGF responsiveness. FASEB J

15:447–457. doi:10.1096/fj.00-0139com

68. Fiedler U, Reiss Y, Scharpfenecker M et al (2006) Angiopoietin-

2 sensitizes endothelial cells to TNF-alpha and has a crucial role

in the induction of inflammation. Nat Med 12:235–239. doi:

10.1038/nm1351

69. Shimoda H, Bernas MJ, Witte MH et al (2007) Abnormal

recruitment of periendothelial cells to lymphatic capillaries in

digestive organs of angiopoietin-2-deficient mice. Cell Tissue

Res 328:329–337. doi:10.1007/s00441-006-0360-8

70. Hackett SF, Wiegand S, Yancopoulos G et al (2002) Angio-

poietin-2 plays an important role in retinal angiogenesis. J Cell

Physiol 192:182–187. doi:10.1002/jcp.10128

71. Dumont DJ, Gradwohl GJ, Fong GH et al (1993) The endo-

thelial-specific receptor tyrosine kinase, tek, is a member of a

new subfamily of receptors. Oncogene 8:1293–1301

72. Kim I, Kim HG, So JN et al (2000) Angiopoietin-1 regulates

endothelial cell survival through the phosphatidylinositol 3’-

Kinase/Akt signal transduction pathway. Circ Res 86:24–29

73. Jones N, Iljin K, Dumont DJ et al (2001) Tie receptors: new

modulators of angiogenic and lymphangiogenic responses. Nat

Rev Mol Cell Biol 2:257–267. doi:10.1038/35067005

74. Hodous BL, Geuns-Meyer SD, Hughes PE et al (2007) Evolu-

tion of a highly selective and potent 2-(pyridin-2-yl)-1, 3, 5-

triazine Tie-2 kinase inhibitor. J Med Chem 50:611–626. doi:

10.1021/jm061107l

75. Semones M, Feng Y, Johnson N et al (2007) Pyridinylimidazole

inhibitors of Tie2 kinase. Bioorg Med Chem Lett 17:4756–4760.

doi:10.1016/j.bmcl.2007.06.068

76. Cardone MH, Roy N, Stennicke HR et al (1998) Regulation of

cell death protease caspase-9 by phosphorylation. Science

282:1318–1321. doi:10.1126/science.282.5392.1318

77. Papapetropoulos FultonD, Mahboubi KA et al (2000) Angio-

poietin-1 inhibits endothelial cell apoptosis via the Akt/survivin

pathway. J Biol Chem 275:9102–9105. doi:10.1074/jbc.275.13.

9102

78. Harfouche R, Hassessian HM, Guo Y et al (2002) Mechanisms

which mediate the antiapoptotic effects of angiopoietin-1 on

endothelial cells. Microvasc Res 64:135–147. doi:10.1006/mvre.

2002.2421

79. Iivanainen E, Nelimarkka L, Elenius V et al (2003) Angio-

poietin-regulated recruitment of vascular smooth muscle cells

by endothelial-derived heparin binding EGF-like growth factor.

FASEB J 17:1609–1621. doi:10.1096/fj.02-0939com

80. Kobayashi H, DeBusk LM, Babichev YO et al (2006) Hepato-

cyte growth factor mediates angiopoietin-induced smooth mus-

cle cell recruitment. Blood 108:1260–1266. doi:10.1182/blood-

2005-09-012807

81. Lindahl P, Johansson BR, Leveen P et al (1997) Pericyte loss

and microaneurysm formation in PDGF-B-deficient mice. Sci-

ence 277:242–245. doi:10.1126/science.277.5323.242

82. Hellstrom M, Kalen M, Lindahl P et al (1999) Role of PDGF-B

and PDGFR-beta in recruitment of vascular smooth muscle cells

and pericytes during embryonic blood vessel formation in the

mouse. Development 126:3047–3055

83. Uemura A et al (2002) Recombinant angiopoietin-1 restores

higher-order architecture of growing blood vessels in mice in the

absence of mural cells. J Clin Invest 110:1619–1628

84. Hirschi KK, Rohovsky SA, D’Amore PA (1998) PDGF, TGF-

beta, and heterotypic cell–cell interactions mediate endothelial

cell-induced recruitment of 10T1/2 cells and their differentiation

to a smooth muscle fate. J Cell Biol 141:805–814. doi:

10.1083/jcb.141.3.805

85. Oh SP, Seki T, Goss KA et al (2000) Activin receptor-like

kinase 1 modulates transforming growth factor-beta 1 signaling

in the regulation of angiogenesis. Proc Natl Acad Sci USA

97:2626–2631. doi:10.1073/pnas.97.6.2626

86. Nishishita T, Lin PC (2004) Angiopoietin 1, PDGF-B, and TGF-

beta gene regulation in endothelial cell and smooth muscle cell

interaction. J Cell Biochem 91:584–593. doi:10.1002/jcb.10718

87. Kidoya H, Ueno M, Yamada Y et al (2008) Spatial and temporal

role of the apelin/APJ system in the caliber size regulation of

blood vessels during angiogenesis. EMBO J 27:522–534. doi:

10.1038/sj.emboj.7601982

88. Fukuhara S, Sako K, Minami T et al (2008) Differential function

of Tie2 at cell-cell contacts and cell-substratum contacts

Angiogenesis (2009) 12:125–137 135

123

Page 12: The role of the Angiopoietins in vascular morphogenesis

regulated by angiopoietin-1. Nat Cell Biol 10:513–526. doi:

10.1038/ncb1714

89. Saharinen P, Eklund L, Miettinen J et al (2008) Angiopoietins

assemble distinct Tie2 signalling complexes in endothelial cell–

cell and cell-matrix contacts. Nat Cell Biol 10:527–537. doi:

10.1038/ncb1715

90. Jones N, Dumont DJ (1998) The Tek/Tie2 receptor signals

through a novel Dok-related docking protein, Dok-R. Oncogene

17:1097–1108. doi:10.1038/sj.onc.1202115

91. Gavard J, Gutkind JS (2006) VEGF controls endothelial-cell

permeability by promoting the beta-arrestin-dependent endocy-

tosis of VE-cadherin. Nat Cell Biol 8:1223–1234. doi:10.1038/

ncb1486

92. Dejana E, Orsenigo F, Lampugnani MG (2008) The role of

adherens junctions and VE-cadherin in the control of vascular

permeability. J Cell Sci 121:2115–2122. doi:10.1242/jcs.017897

93. Gavard J, Patel V, Gutkind JS (2008) Angiopoietin-1 prevents

VEGF-induced endothelial permeability by sequestering Src

through mDia. Dev Cell 14:25–36. doi:10.1016/j.devcel.2007.

10.019

94. Li X, Hahn CN, Parsons M et al (2004) Role of protein kinase

Czeta in thrombin-induced endothelial permeability changes:

inhibition by angiopoietin-1. Blood 104:1716–1724. doi:

10.1182/blood-2003-11-3744

95. Li X, Stankovic M, Bonder CS et al (2008) Basal and angio-

poietin-1-mediated endothelial permeability is regulated by

sphingosine kinase-1. Blood 111:3489–3497. doi:10.1182/

blood-2007-05-092148

96. Huang L, Turck CW, Rao P et al (1995) GRB2 and SH-PTP2:

potentially important endothelial signaling molecules down-

stream of the TEK/TIE2 receptor tyrosine kinase. Oncogene

11:2097–2103

97. Kontos CD, Stauffer TP, Yang WP et al (1998) Tyrosine 1101 of

Tie2 is the major site of association of p85 and is required for

activation of phosphatidylinositol 3-kinase and Akt. Mol Cell

Biol 18:4131–4140

98. Peters KG, Kontos CD, Lin PC et al (2004) Functional signifi-

cance of Tie2 signaling in the adult vasculature. Recent Prog

Horm Res 59:51–71. doi:10.1210/rp.59.1.51

99. Fujikawa K, de Aos Scherpenseel I, Jain SK et al (1999) Role of

PI 3-kinase in angiopoietin-1-mediated migration and attach-

ment-dependent survival of endothelial cells. Exp Cell Res

253:663–672. doi:10.1006/excr.1999.4693

100. Kim I, Oh JL, Ryu YS et al (2002) Angiopoietin-1 negatively

regulates expression and activity of tissue factor in endothelial

cells. FASEB J 16:126–128

101. Zhu WH, Nicosia RF (2002) The thin prep rat aortic ring assay:

a modified method for the characterization of angiogenesis in

whole mounts. Angiogenesis 5:81–86. doi:10.1023/A:1021509

004829

102. Jones N, Chen SH, Sturk C et al (2003) A unique autophos-

phorylation site on Tie2/Tek mediates Dok-R phosphotyrosine

binding domain binding and function. Mol Cell Biol 23:2658–

2668. doi:10.1128/MCB.23.8.2658-2668.2003

103. Kim I, Kim HG, Moon SO et al (2000) Angiopoietin-1 induces

endothelial cell sprouting through the activation of focal adhe-

sion kinase and plasmin secretion. Circ Res 86:952–959

104. Tournaire R, Simon MP, le Noble F et al (2004) A short syn-

thetic peptide inhibits signal transduction, migration and angi-

ogenesis mediated by Tie2 receptor. EMBO Rep 5:262–267.

doi:10.1038/sj.embor.7400100

105. Witzenbichler B, Westermann D, Knueppel S et al (2005)

Protective role of angiopoietin-1 in endotoxic shock. Circulation

111:97–105. doi:10.1161/01.CIR.0000151287.08202.8E

106. Kim I, Moon SO, Park SK et al (2001) Angiopoietin-1 reduces

VEGF-stimulated leukocyte adhesion to endothelial cells by

reducing ICAM-1, VCAM-1, and E-selectin expression. Circ

Res 89:477–479. doi:10.1161/hh1801.097034

107. Nykanen AI, Krebs R, Saaristo A et al (2003) Angiopoietin-1

protects against the development of cardiac allograft arterio-

sclerosis. Circulation 107:1308–1314. doi:10.1161/01.CIR.00

00054623.35669.3F

108. Cho CH, Kammerer RA, Lee HJ et al (2004) Designed angio-

poietin-1 variant, COMP-Ang1, protects against radiation-

induced endothelial cell apoptosis. Proc Natl Acad Sci USA

101:5553–5558. doi:10.1073/pnas.0307575101

109. Hughes DP, Marron MB, Brindle NP (2003) The antiinflam-

matory endothelial tyrosine kinase Tie2 interacts with a novel

nuclear factor-kappaB inhibitor ABIN-2. Circ Res 92:630–636.

doi:10.1161/01.RES.0000063422.38690.DC

110. Jeon BH, Khanday F, Deshpande S et al (2003) Tie-ing the

antiinflammatory effect of angiopoietin-1 to inhibition of NF-

kappaB. Circ Res 92:586–588. doi:10.1161/01.RES.00000

66881.04116.45

111. Tadros A, Hughes DP, Dunmore BJ et al (2003) ABIN-2 pro-

tects endothelial cells from death and has a role in the antiap-

optotic effect of angiopoietin-1. Blood 102:4407–4409. doi:

10.1182/blood-2003-05-1602

112. Gravallese EM, Pettit AR, Lee R et al (2003) Angiopoietin-1 is

expressed in the synovium of patients with rheumatoid arthritis

and is induced by tumour necrosis factor alpha. Ann Rheum Dis

62:100–107. doi:10.1136/ard.62.2.100

113. Scott BB, Zaratin PF, Gilmartin AG et al (2005) TNF-alpha

modulates angiopoietin-1 expression in rheumatoid synovial

fibroblasts via the NF-kappa B signalling pathway. Biochem

Biophys Res Commun 328:409–414. doi:10.1016/j.bbrc.2004.12.180

114. Grall F, Gu X, Tan L et al (2003) Responses to the proinflam-

matory cytokines interleukin-1 and tumor necrosis factor alpha

in cells derived from rheumatoid synovium and other joint tis-

sues involve nuclear factor kappaB-mediated induction of the

Ets transcription factor ESE-1. Arthritis Rheum 48:1249–1260.

doi:10.1002/art.10942

115. Brown C, Gaspar J, Pettit A et al (2004) ESE-1 is a novel

transcriptional mediator of angiopoietin-1 expression in the

setting of inflammation. J Biol Chem 279:12794–12803. doi:

10.1074/jbc.M308593200

116. Parikh SM, Mammoto T, Schultz A et al (2006) Excess circu-

lating angiopoietin-2 may contribute to pulmonary vascular leak

in sepsis in humans. PLoS Med 3:e46. doi:10.1371/journal.

pmed.0030046

117. Gallagher DC, Parikh SM, Balonov K et al (2007) Circulating

Angiopoietin 2 correlates with mortality in a surgical population

with acute lung injury/adult respiratory distress syndrome.

Shock 29:656–661

118. Orfanos SE, Kotanidou A, Glynos C et al (2007) Angiopoietin-2

is increased in severe sepsis: correlation with inflammatory

mediators. Crit Care Med 35:199–206. doi:10.1097/01.CCM.

0000251640.77679.D7

119. Siner JM, Bhandari V, Engle KM, et al. (2009) Elevated serum

Angiopoietin 2 levels are associated with increased mortality in

sepsis. Shock 31:348–353

120. Fearon U, Griosios K, Fraser A et al (2003) Angiopoietins,

growth factors, and vascular morphology in early arthritis. J

Rheumatol 30:260–268

121. Kuroda K, Sapadin A, Shoji T et al (2001) Altered expression of

angiopoietins and Tie2 endothelium receptor in psoriasis. J

Invest Dermatol 116:713–720. doi:10.1046/j.1523-1747.2001.

01316.x

122. Etoh T, Inoue H, Tanaka S et al (2001) Angiopoietin-2 is related

to tumor angiogenesis in gastric carcinoma: possible in vivo

regulation via induction of proteases. Cancer Res 61:2145–2153

136 Angiogenesis (2009) 12:125–137

123

Page 13: The role of the Angiopoietins in vascular morphogenesis

123. Hu B, Guo P, Fang Q et al (2003) Angiopoietin-2 induces

human glioma invasion through the activation of matrix metal-

loprotease-2. Proc Natl Acad Sci USA 100:8904–8909. doi:

10.1073/pnas.1533394100

124. Mandriota SJ, Pepper MS (1998) Regulation of angiopoietin-2

mRNA levels in bovine microvascular endothelial cells by

cytokines and hypoxia. Circ Res 83:852–859

125. Krikun G, Schatz F, Finlay T et al (2000) Expression of an-

giopoietin-2 by human endometrial endothelial cells: regulation

by hypoxia and inflammation. Biochem Biophys Res Commun

275:159–163. doi:10.1006/bbrc.2000.3277

126. Yamakawa M, Liu LX, Date T et al (2003) Hypoxia-inducible

factor-1 mediates activation of cultured vascular endothelial

cells by inducing multiple angiogenic factors. Circ Res 93:664–

673. doi:10.1161/01.RES.0000093984.48643.D7

127. Hegen A, Koidl S, Weindel K et al (2004) Expression of angio-

poietin-2 in endothelial cells is controlled by positive and negative

regulatory promoter elements. Arterioscler Thromb Vasc Biol

24:1803–1809. doi:10.1161/01.ATV.0000140819.81839.0e

128. Lund EL, Hog A, Olsen MW et al (2004) Differential regulation

of VEGF, HIF1alpha and angiopoietin-1, -2 and -4 by hypoxia

and ionizing radiation in human glioblastoma. Int J Cancer

108:833–838. doi:10.1002/ijc.11662

129. Watanabe D, Takagi H, Suzuma K et al (2004) Transcription

factor Ets-1 mediates ischemia- and vascular endothelial growth

factor-dependent retinal neovascularization. Am J Pathol

164:1827–1835

130. Goettsch W, Gryczka C, Korff T et al (2008) Flow-dependent

regulation of angiopoietin-2. J Cell Physiol 214:491–503. doi:

10.1002/jcp.21229

131. Milkiewicz M, Uchida C, Gee E et al. (2008) Shear stress-

induced Ets-1 modulates protease inhibitor expression in

microvascular endothelial cells. J Cell Physiol

132. Daly C, Wong V, Burova E et al (2004) Angiopoietin-1 mod-

ulates endothelial cell function and gene expression via the

transcription factor FKHR (FOXO1). Genes Dev 18:1060–1071.

doi:10.1101/gad.1189704

133. Vajkoczy P, Farhadi M, Gaumann A et al (2002) Microtumor

growth initiates angiogenic sprouting with simultaneous

expression of VEGF, VEGF receptor-2, and angiopoietin-2. J

Clin Invest 109:777–785

134. Machein MR, Knedla A, Knoth R et al (2004) Angiopoietin-1

promotes tumor angiogenesis in a rat glioma model. Am J Pathol

165:1557–1570

135. Zhou YZ, Fang XQ, Li HQ et al (2007) Role of serum angio-

poietin-2 level in screening for esophageal squamous cell cancer

and its precursors. Chin Med J (Engl) 120:1216–1219

136. Kuboki S, Shimizu H, Mitsuhashi N et al. (2008) Angiopoietin-2

levels in the hepatic vein as a useful predictor of tumor inva-

siveness and prognosis in human hepatocellular carcinoma. J

Gastroenterol Hepatol 23:157–164

137. Park JH, Park KJ, Kim YS et al (2007) Serum angiopoietin-2 as

a clinical marker for lung cancer. Chest 132:200–206. doi:

10.1378/chest.06-2915

138. Ahmad SA, Liu W, Jung YD et al (2001) Differential expression

of angiopoietin-1 and angiopoietin-2 in colon carcinoma. A

possible mechanism for the initiation of angiogenesis. Cancer

92:1138–1143. doi:10.1002/1097-0142(20010901)92:5\1138::

AID-CNCR1431[3.0.CO;2-L

139. Oka N, Yamamoto Y, Takahashi M et al (2005) Expression of

angiopoietin-1 and -2, and its clinical significance in human

bladder cancer. BJU Int 95:660–663. doi:10.1111/j.1464-410X.

2005.05358.x

140. Takanami I (2004) Overexpression of Ang-2 mRNA in non-

small cell lung cancer: association with angiogenesis and poor

prognosis. Oncol Rep 12:849–853

141. Cai M, Zhang H, Hui R (2003) Single chain Fv antibody against

angiopoietin-2 inhibits VEGF-induced endothelial cell prolifer-

ation and migration in vitro. Biochem Biophys Res Commun

309:946–951. doi:10.1016/j.bbrc.2003.08.086

142. White RR, Shan S, Rusconi CP et al (2003) Inhibition of rat

corneal angiogenesis by a nuclease-resistant RNA aptamer

specific for angiopoietin-2. Proc Natl Acad Sci USA 100:5028–

5033. doi:10.1073/pnas.0831159100

143. Imanishi Y, Hu B, Jarzynka MJ et al (2007) Angiopoietin-2

stimulates breast cancer metastasis through the alpha(5)beta(1)

integrin-mediated pathway. Cancer Res 67:4254–4263

144. Yu Q, Stamenkovic I (2001) Angiopoietin-2 is implicated in the

regulation of tumor angiogenesis. Am J Pathol 158:563–570

145. Hawighorst T, Skobe M, Streit M et al (2002) Activation of the

tie2 receptor by angiopoietin-1 enhances tumor vessel matura-

tion and impairs squamous cell carcinoma growth. Am J Pathol

160:1381–1392

146. Hayes AJ, Huang WQ, Yu J et al (2000) Expression and func-

tion of angiopoietin-1 in breast cancer. Br J Cancer 83:1154–

1160. doi:10.1054/bjoc.2000.1437

147. Stoeltzing O, Ahmad SA, Liu W et al (2003) Angiopoietin-1

inhibits vascular permeability, angiogenesis, and growth of

hepatic colon cancer tumors. Cancer Res 63:3370–3377

148. Tian S, Hayes AJ, Metheny-Barlow LJ et al (2002) Stabilization

of breast cancer xenograft tumour neovasculature by angio-

poietin-1. Br J Cancer 86:645–651. doi:10.1038/sj.bjc.6600082

149. Nakayama T, Yao L, Tosato G (2004) Mast cell-derived an-

giopoietin-1 plays a critical role in the growth of plasma cell

tumors. J Clin Invest 114:1317–1325

150. Shim WS, Teh M, Mack PO et al (2001) Inhibition of angio-

poietin-1 expression in tumor cells by an antisense RNA

approach inhibited xenograft tumor growth in immunodeficient

mice. Int J Cancer 94:6–15. doi:10.1002/ijc.1428

151. Carlson TR, Feng Y, Maisonpierre PC et al (2001) Direct cell

adhesion to the angiopoietins mediated by integrins. J Biol

Chem 276:26516–26525. doi:10.1074/jbc.M100282200

152. Hu B, Jarzynka MJ, Guo P et al (2006) Angiopoietin 2 induces

glioma cell invasion by stimulating matrix metalloprotease 2

expression through the alphavbeta1 integrin and focal adhesion

kinase signaling pathway. Cancer Res 66:775–783. doi:

10.1158/0008-5472.CAN-05-1149

153. Cascone I, Napione L, Maniero F et al (2005) Stable interaction

between alpha5beta1 integrin and Tie2 tyrosine kinase receptor

regulates endothelial cell response to Ang-1. J Cell Biol

170:993–1004. doi:10.1083/jcb.200507082

154. Dallabrida SM, Ismail N, Oberle JR et al (2005) Angiopoietin-1

promotes cardiac and skeletal myocyte survival through inte-

grins. Circ Res 96:e8–e24. doi:10.1161/01.RES.0000158285.

57191.60

155. Dallabrida SM, Ismail NS, Pravda EA et al (2008) Integrin

binding angiopoietin-1 monomers reduce cardiac hypertrophy.

FASEB J 22:3010–3023. doi:10.1096/fj.07-100966

156. Ward NL, Putoczki T, Mearow K et al (2005) Vascular-specific

growth factor angiopoietin 1 is involved in the organization of

neuronal processes. J Comp Neurol 482:244–256. doi:10.1002/

cne.20422

157. Valable S, Bellail A, Lesne S et al (2003) Angiopoietin-1-

induced PI3-kinase activation prevents neuronal apoptosis.

FASEB J 17:443–445

158. Arai F, Hirao A, Ohmura M et al (2004) Tie2/angiopoietin-1

signaling regulates hematopoietic stem cell quiescence in the

bone marrow niche. Cell 118:149–161. doi:10.1016/j.cell.2004.

07.004

Angiogenesis (2009) 12:125–137 137

123