Вы находитесь на странице: 1из 14

Downloaded from http://perspectivesinmedicine.cshlp.

org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

The Complex Role of Angiopoietin-2 in the


Angiopoietin Tie Signaling Pathway

Gavin Thurston and Christopher Daly


Regeneron Pharmaceuticals, Tarrytown, New York 10591
Correspondence: gavin.thurston@regeneron.com

The angiopoietin Tie signaling system is a vascular-specific receptor tyrosine kinase


pathway that is essential for normal vascular development. Although the basic functioning
of the pathway is understood, many uncertainties remain about the role of certain
members of the pathway, particularly angiopoietin-2 (Ang2), in pathological vascular remod-
eling and angiogenesis. We summarize the components of the angiopoietinTie pathway
and then focus on studies that highlight the role of Ang2 in disease settings, including
cancer and inflammation. The expression of Ang2 is elevated in many cancers and types
of inflammation, which prompted the development of specific reagents to block its inter-
action with the Tie2 receptor. The application of these reagents in preclinical models of
inflammation and cancer has begun to elucidate the role of Ang2 in vascular remodeling
and disease pathogenesis and has led to emerging clinical tests of Ang2 inhibitors.
www.perspectivesinmedicine.org

he angiopoietin Tie signaling system was The core components of the signaling sys-
T identified as a vascular-specific receptor
tyrosine kinase pathway that is essential for
tem appear to be angiopoietin-1 (Ang1) and
Tie2, in that Ang1 is a definitive activating ago-
vessel development. This signaling system has nist of the pathway and Tie2 is the cognate
many important parallels to the better under- receptor. However, perhaps because of their
stood VEGF system. For example, the Tie recep- regulated expression patterns, other members
tors (Tie1 and Tie2, or Tek) are expressed of the pathway have emerged as desirable thera-
selectively by endothelial cells, similar to what peutic targets for drug development. For exam-
has been found with VEGF receptors. Signaling ple, several approaches have been developed to
by Tie receptors appears to complement the selectively block Ang2. Despite much research
VEGF pathway by contributing to later stages over the past decade, our understanding of the
of vascular development. Thus, whereas VEGF role of Ang2 in the angiopoietin Tie signaling
signals promote initiating events in angio- system, and vascular biology in general, is
genesis such as endothelial cell sprouting, particularly murky. For example, it is still
angiopoietin Tie signals appear to promote unclear whether Ang2 is an antagonist or ago-
endothelial cell survival and vascular assembly, nist of Tie2 in settings of vascular remodeling.
stability, and maturation. Increased understanding of Ang2 will become

Editors: Michael Klagsbrun and Patricia DAmore


Additional Perspectives on Angiogenesis available at www.perspectivesinmedicine.org
Copyright # 2012 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a006650
Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650

1
Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

G. Thurston and C. Daly

especially important as these inhibitors move Ang1 Ang2


forward in the clinic and are tested in combina-
tion with other anti-angiogenic agents. This
work summarizes the components and basic
biology of the angiopoietin Tie pathway, de-
scribes in more detail studies that reveal the
increased expression of Ang2 in human disease
as well as mechanistic studies that shed light on
its role in preclinical disease models, and then
attempts to highlight the outstanding questions
for our understanding of the role of Ang2 in
angiopoietin Tie2 signaling and vascular biol-
ogy. For a more general summary of the angio-
poietin Tie pathway, the reader is directed to
an excellent recent review (Augustin et al. 2009).

BASIC BIOLOGY OF THE ANGIOPOIETIN


Tie2 PATHWAY Tie1 Tie2 VE-PTP

ReceptorsTie1 and Tie2 Figure 1. Molecular components of the angiopoie-


The receptors Tie1 and Tie2 are expressed selec- tin Tie pathway. The multimeric ligands Ang1 and
Ang2 bind to Tie2 receptor. Tie1 receptor can interact
tively by endothelial cells, although other cell
www.perspectivesinmedicine.org

with Tie2, although it apparently does not bind


types including early hematopoietic cells and directly to Ang1 or Ang2. The receptor tyrosine phos-
subsets of monocytes also express Tie2. Despite phatase VE-PTP dephosphorylates Tie2. In the
a high degree of structural homology, the two extracellular regions of receptors: (blue circles) Ig-like
receptors have markedly different properties domains; (green boxes) fibronectin type III domains;
(Sato et al. 1993; Augustin et al. 2009). Structur- (red boxes) EGF-like domains. Components are not
ally, in the extracellular portion, both receptors drawn to scale.
are composed of two immunoglobulin (Ig)
like domains, followed by three EGF-like do- Knockdown of Tie1 by siRNA indicates that
mains, another Ig-like domain, and three Tie1 is not required for Ang1-dependent activa-
fibronectin type III domains (Fig. 1). In the tion of the AKT or ERK pathways (Yuan et al.
cytoplasmic portion, both Tie1 and Tie2 con- 2007). Thus, the functional role of Tie1 in
tain split tyrosine kinase domains. angiopoietin signaling remains unclear.
Functionally, Tie2 binds directly to angio- Genetic deletion of Tie2 confirms its role as
poietins and has strong kinase activity. In the core signaling component, because mice
contrast, Tie1 does not bind directly to angio- null for Tie2 exhibit severe vascular and cardiac
poietins under normal conditions and has abnormalities that lead to embryonic lethality at
weak kinase activity. Following binding to approximately embryonic day 10.5 (E10.5)
Ang1, Tie2 becomes phosphorylated on several (Dumont et al. 1994; Sato et al. 1995). In com-
cytoplasmic tyrosine residues, which results in parison, genetic deletion of Tie1 leads to vascu-
activation of downstream signaling pathways lar perturbation later in development and
including the PI3-kinase/AKT and ERK path- embryonic lethality that is somewhat variable
ways. In comparison, although Tie1 does in onset (E13.5 to birth) (Puri et al. 1995;
not directly bind to angiopoietins, it forms a Sato et al. 1995).
complex with angiopoietins and Tie2 and Important insights have come from work
also becomes phosphorylated on cytoplasmic linking human venous malformations to muta-
tyrosine residues (Saharinen et al. 2005). tions in the Tie2 gene. Initially, heritable venous

2 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

Angiopoietin Tie Signaling Pathway

malformations in two families were found to be expressed by pericytes and perivascular cells in
associated with a missense mutation in the ki- normal vessels. Its expression is not strongly
nase domain of Tie2 (Vikkula et al. 1996). changed by most vascular stimuli. Interestingly,
This mutation results in increased activity of Ang1 is stored at high levels in platelet granules
Tie2. Subsequent studies have found that spora- (Li et al. 2001).
dic venous malformations are also associated Ang2 was initially identified by homology
with somatic point mutations in Tie2, which with Ang1 (Maisonpierre et al. 1997). Ang2
again are activating (Limaye et al. 2009). was found to bind to Tie2 with a similar affinity
as Ang1. However, unlike Ang1, exogenous
Ang2 provided only a very weak activation of
LigandsAngiopoietin-1 and Angiopoietin-2
Tie2 on endothelial cells. When exogenous
Angiopoietins are secreted, multimeric ligands. Ang2 and Ang1 were added together, the levels
There are three genuine angiopoietins (Ang1, of Tie2 phosphorylation were decreased com-
-2, and -4 in human) that bind to Tie2, plus sev- pared with addition of Ang1 alone. This and
eral angiopoietin-like molecules that share the other results (see below) led to the model that
same structure and have sequence homology Ang2 was an antagonist of Tie2 (Hanahan
to the angiopoietins but do not bind Tie recep- 1997; Maisonpierre et al. 1997).
tors. Ang1 and Ang2 have been the most exten- Ang2 is expressed predominantly by endo-
sively studied and are described here (Fig. 1), thelial cells and some smooth muscle cells.
whereas Ang4 has been much less studied and Unlike Ang1, the expression of Ang2 is strongly
does not yet have a clearly defined role in phys- regulated: Its expression is normally low in qui-
iology or pathology. escent mature vessels but is strongly increased in
Angiopoietin proteins are comprised of an many inflammatory and angiogenic settings.
www.perspectivesinmedicine.org

amino-terminal domain that serves to promote For example, Ang2 expression in cultured endo-
higher-order clustering of the molecules, fol- thelial cells is increased by TNF, VEGF, and
lowed by a coiled-coil domain that promotes hypoxia (Mandriota and Pepper 1998; Kim
multimerization, and a carboxy-terminal fibri- et al. 2000c). In the ovary of mature female
nogen homology domain that contains the rats, which is a site of physiologic angiogenesis
binding sites for Tie2 (Davis et al. 2003). Angio- and vascular remodeling, the expression of
poietins form homomeric higher-order multi- Ang2 was shown to have dramatic variations
mers containing from three to six (or more) during the course of the follicular cycle, which
individual proteins. In general, they are rather mirror similar dramatic changes in expression
sticky proteins, particularly Ang1, with re- of VEGF (Maisonpierre et al. 1997).
ported interactions with extracellular matrix In cultured endothelial cells, Ang2 protein is
(Xu and Yu 2001). As a result, much effort has often stored in intracellular granules (Fiedler
been made to generate artificial recombinant et al. 2004). These granules can release Ang2
forms of Ang1 (Davis et al. 2003; Cho et al. in response to stimuli such as PMA. However,
2004). more documentation is needed as to whether
Angiopoietin-1 was initially identified as an normal quiescent or angiogenic endothelial
activating ligand for Tie2 that is expressed by cells in vivo store Ang2 protein in granules.
perivascular cells (Davis et al. 1996). Genetic Regardless of whether Ang2 is stored in granules
deletion of Ang1 results in embryonic lethality, or not, it is clearly secreted by activated endo-
with severe heart and vascular defects, very sim- thelium and has been shown to be elevated in
ilar in phenotype to Tie2-null mice, and death the plasma in a variety of inflammatory and
at E12.5 (Suri et al. 1996). The blood vessels angiogenic diseases, including sepsis, malaria,
in Ang1-null mice form but lack proper attach- and cancer (see below).
ment of pericytes and fail to mature. Ang1 is Genetic manipulation of Ang2 in mice has
strongly expressed in the heart during mid- validated its role in vascular development but
gestational development and is moderately has also revealed its complexity. The initial

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650 3


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

G. Thurston and C. Daly

studies with genetic manipulation described activity of VE-PTP has been linked to its
transgenic overexpression of Ang2 in endothe- dephosphorylation of Tie2 (Li et al. 2009;
lial cells (via a Tie2 promoter) (Maisonpierre Winderlich et al. 2009) and subsequent down-
et al. 1997). No viable mice were found, but, regulation of ERK signaling. Thus, VE-PTP
rather, this manipulation yielded embryonic appears to be an essential negative regulator of
lethality at E9.5 E10. Embryos analyzed at angiopoietin Tie signaling. It is worth noting
E9 E9.5 showed dramatic disruption of the that the reported phenotype of the VE-PTP
vasculature and heart. The effects of overexpres- knockout mice, which is likely mediated by
sion of Ang2 were described as similar to those overactivation of Tie2, is somewhat similar to
seen in embryos lacking either Tie2 or Ang1, that of the Ang1 and Tie2 knockout mice,
thus providing evidence that Ang2 was acting making the interpretation of the Ang2-over-
as an antagonist for Tie2. expressing mice more difficult. A more detailed
Mice genetically deficient for Ang2 showed and contemporaneous examination of these
complex lymphatic and vascular phenotypes multiple genetic manipulations would be
(Gale et al. 2002). Knockout (KO) mice were valuable.
born at approximately normal Mendelian ratios
but soon developed chylous ascites and edema,
Cellular and Vascular Effects of Angiopoietin
and most mice died within the first 2 wk postna-
Tie Signaling
tally. An underlying defect was found in the cen-
tral lacteals of the intestinal villi and throughout Several studies in mice and with cultured endo-
the lymphatic system, which likely accounted thelial cells suggest that Ang1 promotes vessel
for the perinatal lethality. In addition, defects stabilization and enlargement without inducing
were found in the early postnatal remodeling vascular sprouting. Genetic overexpression of
www.perspectivesinmedicine.org

of the vasculature in the eye: The hyaloid vessels Ang1 in the skin of mice resulted in enlarged
failed to regress properly, and the retinal vessels dermal vessels (Suri et al. 1998; Thurston et al.
grew abnormally. Subtle defects were also noted 1999). Treatment of normal mice with exoge-
in the patterning of the vasculature in several nous Ang1 protein can also induce vascular
organs, although the mechanism for these enlargement, associated with an increased
defects was not determined. number of endothelial cells (Baffert et al. 2004;
Thurston et al. 2005). Importantly, vessels
exposed to Ang1 are resistant to leak induced
Receptor PhosphataseVE-PTP
by VEGF or inflammatory agents (Thurston
The angiopoietin Tie pathway appears to have et al. 1999, 2000). In addition to its anti-leakage
another key player, namely, the receptor tyro- effects, acute exposure of vasculature to Ang1
sine phosphatase VE-PTP (also called PTPRb). can reduce vascular activation and dysfunction.
VE-PTP is an endothelial cell-specific trans- For example, Ang1 inhibits leukocyte infiltra-
membrane phosphatase that consists of a large tion in models of diabetic retinopathy (Joussen
extracellular domain with 17 fibronectin type et al. 2002) and sepsis (Witzenbichler et al.
III repeats, a transmembrane domain, and a 2005). These results suggest a role for Ang1
cytoplasmic phosphatase domain (Fig. 1). Tie2 signaling in maintaining a quiescent, well-
Genetic inactivation of VE-PTP in mice results functioning vasculature.
in embryonic lethality, with defects in vascular The effects of Ang1 on leukocyte infiltration
remodeling and heart development (Baumer in inflammatory models may reflect, at least
et al. 2006; Dominguez et al. 2007). The extra- partly, inhibition of endothelial cell adhesion
cellular domain of VE-PTP appears to be im- molecule expression. Decreased expression of
portant for its function because antibodies to adhesion proteins including ICAM-1, VCAM-
this region can cause defects in vascular remod- 1, and, E-selectin has been attributed to activa-
eling similar to those seen with genetic inactiva- tion of the PI3-kinase/AKT pathway (Kim et al.
tion of VE-PTP (Winderlich et al. 2009). The 2001). With respect to permeability, several

4 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

Angiopoietin Tie Signaling Pathway

reports suggest that Ang1 works by inhibiting cultured endothelial cells by low AKT signaling
calcium influx and/or activation of the small and the transcription factor FOXO1, and in
GTPase Rho (Li et al. 2004; Jho et al. 2005). this setting Ang2 was shown to activate Tie2
Both calcium flux and Rho are believed to pro- (Daly et al. 2006). Tie2 activation in this model
mote permeability through effects on the actin was blocked by antibodies to Ang2, indicating
cytoskeleton. an autocrine or juxtacrine role for Ang2.
The effects of Ang1 Tie2 signaling on the Thus, Ang2 expression and secretion may be a
inflammatory response may be more complex compensatory response by endothelial cells
than described above. A recent study has shown to low AKT activity, for example, when Ang1/
that chronic activation of Tie2 by exogenous Tie2 signaling is weak (Fig. 2). Conversely,
Ang1 in mice can change airway microvessels strong AKT activation in response to Ang1
from a capillary-like phenotype to that of a Tie2 activity leads to inhibition of Ang2 expres-
venule-like phenotype (Fuxe et al. 2010). sion (Daly et al. 2004). These findings suggest a
Such phenotypic changes in the microvascula- model in which endothelial cells that actively
ture also occur in chronic inflammation. produce Ang2 can respond by robust activation
Because the postcapillary venules are typically of Tie2.
the site of the inflammatory response, the In addition to these effects in cultured
expanded venule-like phenotype produced by endothelial cells, Ang1 and Ang2 can produce
Ang1 stimulation produced a more vigorous similar effects in vivo. For example, both Ang1
response to acute inflammatory challenge and Ang2 rapidly activated Tie2/AKT signaling
(Fuxe et al. 2010). The anti-inflammatory in mouse heart and inhibited expression of
effects of Ang1 may depend on the dosage or FOXO1 target genes, including Ang2 itself
duration of exposure to Ang1 and the nature (Daly et al. 2006). Additionally, replacement
www.perspectivesinmedicine.org

of the acute inflammatory stimuli. of the Ang2 gene with a cDNA encoding Ang1
Recent studies indicate that signaling down- led to a partial rescue of the phenotype in Ang2-
stream from Tie2 activation is influenced by the deficient mice (Gale et al. 2002), consistent with
subcellular localization of the receptor, which is an activating role for Ang2. Finally, in several
different in confluent versus sparse endothelial models of inflammation, acute treatment with
cells (Fukuhara et al. 2008; Saharinen et al. either Ang1 or Ang2 can inhibit components
2008). In these studies, Ang1 produced stronger of the inflammatory process (Daly et al. 2006;
AKT signaling in confluent cells, in which Tie2 Nykanen et al. 2006).
was localized at cell/cell junctions. In compari-
son, Ang1 produced stronger ERK signaling
EXPRESSION OF ANG2 IS INCREASED
when the cells were sparse, in which case, Tie2
IN SEVERAL DISEASE SETTINGS
was localized at sites of cell/substratum con-
tacts. Thus, the effects of Ang Tie2 activation Since the initial description of dramatic regula-
on endothelial cell function may depend on tion of Ang2 expression in the cycling ovary,
the nature of the cell cell and cell matrix many studies using a variety of approaches
contacts. have shown that Ang2 is, indeed, highly
As described above, several lines of evidence up-regulated in many diseases and settings of
suggested that Ang2 acts as a Tie2 antagonist. vascular remodeling. Because of a paucity of
However, several other experimental results antibodies to Ang2 that allow robust immuno-
indicate that Ang2 can act as an agonist for histochemistry, very few studies have docu-
Tie2. First, exogenous Ang2 can clearly promote mented the levels or location of Ang2 protein
phosphorylation of Tie2 in cultured endothelial in diseased human tissue. However, several in
cells (Kim et al. 2000b; Teichert-Kuliszewska situ hybridization studies have revealed
et al. 2001), although its effects are not as potent increased levels of Ang2 expression in cancers
as exogenous Ang1. In another model, endoge- of different origin, including neuroendocrine
nous expression of Ang2 was induced in tumors (Detjen et al. 2010), hepatocellular

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650 5


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

G. Thurston and C. Daly

High Ang2 expression


Tumors, inflammation

Low Ang2 expression Ang2


Stable, quiescent endothelium
Autocrine Ang2
Ang1 maintains Tie2
pY pY phosphorylation.
l
na
1 sig
pY pY g ve
An acti Tumor angiogenesis
e ak O
W FOX
PI3K/AKT

FOXO off Inf


la
Cy mm Ang1
tok atio
ine n
s Ang2
Low Ang2 Autocrine or
exogenous Ang2
inhibits Tie2
phosphorylation.

Increased responsiveness to
inflammatory stimuli

Figure 2. Model of agonist and antagonist roles for Ang2 on Tie2 function. In quiescent, mature vessels, Ang1
promotes strong activation of the Tie2/PI3K/AKT pathway. Ang2 expression is low because of AKT-mediated
inhibition of the FOXO1 transcription factor. Low AKTactivity, for example, when Ang1/Tie2 signaling is weak,
www.perspectivesinmedicine.org

results in activation of FOXO1 and increased Ang2 expression. In this setting, Ang2 activates Tie2 phosphory-
lation, thereby compensating for the absence of a strong Ang1 signal. As discussed in the text, some evidence
indicates that Ang2 promotes tumor angiogenesis via activation of Tie2. Expression of Ang2 can also be induced
during inflammation, perhaps by TNFa or other cytokines. In this setting, Ang2 may cause a decrease in Tie2
phosphorylation, by competing with the strong agonist Ang1. Inhibition of Tie2 signaling appears to make the
endothelium more responsive to inflammatory stimuli. It should be stressed that only in very few instances have
changes in Tie2 phosphorylation been directly measured in vivo. Thus, as discussed in the text, models of Ang2
mechanism of action are based largely on indirect evidence.

cancer (Chen et al. 2001; Scholz et al. 2007), gas- A reliable commercial enzyme-linked im-
tric cancer (Moon et al. 2006), angiosarcoma munosorbent assay (ELISA) for human Ang2
(Brown et al. 2000), carcinosarcoma (Emoto has spawned numerous studies that have
et al. 2004), and astrocytoma (Zagzag et al. documented increased levels of Ang2 protein
1999). Interestingly, some studies report in plasma or serum in many disease settings,
increased expression of Ang2 in both endothe- including cancer and inflammation. Using
lial cells and tumor cells (e.g., Helfrich et al. this ELISA, the absolute levels of Ang2 in
2009; Detjen et al. 2010), whereas other studies human plasma reported across a spectrum of
have shown Ang2 expression limited to the vas- studies are reassuringly consistent; plasma
culature and not in tumor cells (Stratmann et al. from healthy controls contains levels of Ang2
1998; Zagzag et al. 1999; Moon et al. 2006; of 1 3 ng/mL, and seldom more than
Goede et al. 2010). In addition to in situ hybrid- 4 ng/mL, whereas plasma from disease shows
ization methods, analyses of RNA from whole a range of Ang2 levels from 5 to 10 ng/mL
and microdissected tissue have shown increased and as high as 20 ng/mL or more. For example,
Ang2 in tumors versus corresponding normal in a study of melanoma, circulating Ang2
tissue (e.g., Durkin et al. 2004; Helfrich et al. levels were increased in patients with Stage III
2009; Goede et al. 2010). and IV disease, with median levels from Stage

6 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

Angiopoietin Tie Signaling Pathway

IV patients almost threefold that of control lev- ANG2 PLAYS A ROLE IN INFLAMMATION-
els and individual values as high as 8 ng/mL INDUCED VASCULAR REMODELING
(Helfrich et al. 2009).
Similar values for circulating Ang2 have As described above, early studies on the pathway
been reported in sepsis. For example, circulat- found that treatment of endothelial cells with
ing levels of Ang2 in severe sepsis (associated recombinant Ang1 reduced the inflammatory
with organ failure) were fivefold increased response to mediators such as VEGF and TNF
compared with normal (Davis et al. 2010). (Gamble et al. 2000; Kim et al. 2001). Similarly,
Another study reported significantly higher exposure of vessels to Ang1 in vivo resulted
levels of circulating Ang2 in patients admitted in vessels that were less leaky in response
for septic shock (van der Heijden et al. 2009). to VEGF and acute inflammatory mediators
Importantly, in this latter study, the levels of (Thurston et al. 2000). These results, combined
Ang2 at admittance were significantly higher with recent data showing increased Ang2 ex-
in nonsurviving patients than in subsequent pression in several inflammatory diseases,
survivors. linked the angiopoietin Tie system to the re-
Recent efforts have made further associa- sponse of endothelial cells to inflammatory
tions between the levels of Ang2 in plasma stimuli. In the past several years, tools to block
and the outcome of the disease. For example, Ang2 function in preclinical disease models
a recent study found that circulating levels of have become available, and several functional
Ang2 were increased in colorectal cancer, with studies have implicated Ang2 more directly in
mean Ang2 levels increasing from 2.3 ng/mL different aspects of inflammation.
in plasma from healthy people to 3.9 ng/mL In addition to abnormalities in the blood
in patients with Stage IV colorectal cancer and lymphatic vasculatures, mice that are genet-
www.perspectivesinmedicine.org

(Goede et al. 2010). Importantly, Ang2 levels ically deficient for Ang2 have deficits in rapid
were much increased in a subset of the patients, leukocyte recruitment to sites of inflammation
with some as high as 12 ng/mL. This report (Fiedler et al. 2006). The response of Ang2 KO
then compared circulating levels of Ang2 with mice to intraperitoneal injection of thioglycol-
outcome ( progression free survival, overall sur- late, which induces rapid accumulation of neu-
vival) in a separate group of treated colorectal trophils, was significantly reduced compared
cancer patients. Using a threshold of 3.5 ng/ with wild-type mice. Ang2 KO mice also showed
mL of circulating Ang2, the study found signifi- reduced recruitment of neutrophils in response
cantly worse outcome for patients with high to intraperitoneal injection of bacteria. The def-
levels of Ang2 compared with those with low icits in leukocyte recruitment in Ang2-deficient
levels (Goede et al. 2010). mice were associated with reduced firm adher-
A study of melanoma also found a correla- ence of neutrophils to vessels after exposure to
tion between high levels of circulating Ang2 TNFa (Fiedler et al. 2006).
and worse outcome (Helfrich et al. 2009). In Another study evaluated the role of Ang2 in a
this study, both progression-free survival and model of acute lung injury in mice (Bhandari
overall survival were reduced in melanoma et al. 2006). Ang2 KO or wild-type mice were
patients with high levels of circulating Ang2 exposed to hyperoxia, which induces lung injury,
(using a threshold of 1.8 ng/mL). Finally, a inflammation, and eventually death. The dura-
study of neuroendocrine tumors found that tion of survival in 100% oxygen was somewhat
the average level of circulating Ang2 increased longer in Ang2 KO mice compared with wild-
from 2.6 ng/mL in control samples to 4.0 ng/ type mice. In addition, mice deficient for Ang2
mL in patients with neuroendocrine tumors, had slightly reduced cellularity and protein levels
with values in the cancer group up to 25 ng/ in bronchio-aveolar lavage (BAL) fluid and less
mL (Detjen et al. 2010). Again, in this tumor alveolar damage as revealed by histology.
type, higher levels of Ang2 were associated A role for Ang2 in inflammation was also
with worse overall survival. demonstrated in a model of airway infection

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650 7


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

G. Thurston and C. Daly

(Tabruyn et al. 2010). Airway inoculation of phenotypic outcomes depending on the context
Mycoplasma pulmonis in mice causes chronic of other cellular and molecular factors. Because
inflammation accompanied by dramatic infil- Ang2 is normally produced in settings of low
tration of leukocytes and remodeling of the Ang1 and/or high levels of activating factors
tracheal microvasculature. Blockade of Ang2 for endothelial cells, the context of Tie2 signal-
during the infection resulted in near-complete ing during times of Ang2 expression could typ-
inhibition of the vascular remodeling and sig- ically be very different from that of quiescent
nificant reduction of infiltrating leukocytes. endothelial cells.
Ang2 blockade also reduced the inflamma-
tory-associated changes in phenotype of the
ANG2 PLAYS A ROLE IN TUMOR
endothelium, such as increased expression of
ANGIOGENESIS AND GROWTH
P- and E-selectin. In this study, the investigators
used antibodies to immunolocalize phosphory- As detailed above, Ang2 is preferentially ex-
lated Tie2 and reported a decrease in phospho- pressed in the endothelial cells of remodeling
Tie2 following Mycoplasma infection (Tabruyn blood vessels, for example, in tumors. Ang2
et al. 2010). Treatment of infected mice with expression has been documented in a range of
Ang2-blocking antibodies resulted in a restora- human cancers, including glioblastoma, mela-
tion of phospho-Tie2, suggesting that Ang2 was noma, prostate adenocarcinoma, and renal cell
contributing to the loss of Tie2 activation. It will carcinoma (see above). The initial model for
be important to validate the phospho-Tie2 Ang2 function in tumors, based on high Ang2
immunohistochemistry with other methods expression in vessels before regression and on
for assessing the levels of Tie2 signaling. Inter- the purported role of Ang2 as a Tie2 antagonist,
estingly, blockade of Ang2 also reduced the was that Ang2-mediated inhibition of Tie2 sig-
www.perspectivesinmedicine.org

overall M. pulmonis load in the airways of naling is required for vessel destabilization,
infected mice compared with mice treated leading to vessel growth in the presence of
with control proteins (Tabruyn et al. 2010). VEGF or to vessel regression in the absence of
This study suggests that Ang2 is a key mediator VEGF (Hanahan 1997; Holash et al. 1999).
of the vascular remodeling and phenotypic Initial studies aimed at defining a functional
changes that occur in chronic inflammation. role for the Tie2 pathway in tumors used a Tie2
In the above settings of inflammation, extracellular domain-Fc fusion protein, which
Ang2 has effects that are seemingly consistent prevents binding of both Ang1 and Ang2 to
with a role as a Tie2 antagonist. Overall, the Tie2. Systemic delivery of soluble Tie2 provided
results suggest that in these models, Ang2 is a modest growth inhibition of mammary tumors
pro-inflammatory factor and helps promote and melanoma in mice, suggesting that active
vessel destabilization (Fig. 2). By extension of signaling through Tie2 is required for robust
previous work suggesting that Ang1 can act to tumor growth (Lin et al. 1998). Because soluble
stabilize vessels and reduce inflammation, the Tie2 blocked both Ang1 and Ang2, these studies
presumption is that Ang2 must be acting via did not shed light on the relative importance of
inhibition of Tie2 signaling (although effects Ang1 versus Ang2, nor did they support the
on Tie2 phosphorylation and downstream sig- model that antagonism of Tie2 activity pro-
naling have not been fully documented). One motes tumor angiogenesis and tumor growth.
explanation for the apparently contradictory A definitive role for Ang2 in tumor growth
findings is that the response of Tie2 to Ang2 was established following the generation of spe-
(either activation or inhibition) is regulated in cific inhibitors (monoclonal antibodies as well
a context-specific fashion by mechanisms that as peptide bodies) that bind to Ang2 and pre-
are not yet understood. However, another vent its interaction with Tie2. Systemic delivery
explanation is that Tie2 activation, by either of Ang2 blockers (which neutralize both mouse
Ang1 or Ang2, does not universally lead to and human Ang2) results in significant, but
vessel stabilization but can produce different partial, inhibition of the growth of a range of

8 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

Angiopoietin Tie Signaling Pathway

human tumor xenografts, including colorec- subcutaneously, embryonic stem cells are ca-
tal, breast, and epidermoid carcinoma (Oliner pable of differentiating into endothelial cells,
et al. 2004; Brown et al. 2010; Huang et al. and they contribute significantly to the vascula-
2010). Although some of the blockers used in ture of the teratoma. Teratomas derived from
these studies inhibit both Ang1 and Ang2, the VE-PTP knockout ES cells have elevated levels
use of completely specific Ang1 and Ang2 of phosphorylated Tie2 and much larger blood
inhibitors has clearly established that Ang2 is vessels than wild-type or VE-PTP heterozygous
the dominant Tie2 ligand in tumors. For exam- tumors, consistent with chronic activation of
ple, an Ang1-specific peptide body has no effect Tie2. Interestingly, treatment of the VE-PTP
on tumor growth and only marginally and knockout tumors with an Ang2-selective anti-
inconsistently potentiates the effect of an Ang2- body decreased Tie2 phosphorylation and ves-
specific peptide body (Falcon et al. 2009; Coxon sel size, suggesting that Ang2 is functioning as
et al. 2010). Similarly, the Ang2-specific peptide a Tie2 activator in this model (Li et al. 2009).
body inhibits tumor growth to approximately This effect of Ang2 blockade on vessel size is
the same extent as a dual anti-Ang1/Ang2 pep- consistent with the data from human tumor
tide body (Oliner et al. 2004; Coxon et al. 2010). xenografts mentioned above that demonstrated
Providing further support for the role of Ang2 decreased vessel size following treatment with
in tumor growth, tumors implanted into Ang2 an Ang2-specific blocker (Falcon et al. 2009).
KO mice grew more slowly than those implanted Further evidence of Ang2 agonist activity
into wild-type mice (Nasarre et al. 2009). comes from the studies cited above that exam-
Consistent with an anti-angiogenic mecha- ined the effect of combined treatment with
nism of action, Ang2 blockers decrease tumor Ang1-specific and Ang2-specific blockers. The
vascularity (Falcon et al. 2009; Brown et al. fact that the Ang1-specific blocker potentiated
www.perspectivesinmedicine.org

2010; Hashizume et al. 2010), apparently via (albeit quite modestly), rather than reversed,
inhibition of vessel sprouting and/or reduction the effects of the Ang2-specific blocker on
of vessel size (Hashizume et al. 2010). Previous tumor growth and tumor vascularity strongly
studies have shown that Tie2 activation causes suggests that these effects of Ang2 blockade
enlargement of blood vessels in normal tissues result from a decrease in Tie2 activity (Falcon
via increased endothelial cell proliferation et al. 2009). Interestingly, the situation becomes
(Thurston et al. 2005). In tumors, Ang2 blockade more complex when the effects of Ang2 block-
significantly inhibits endothelial cell prolifera- ade on vessel normalization are examined in
tion and significantly reduces the size of vessels the same tumors. Although the Ang2-specific
(Falcon et al. 2009), consistent with a decrease blocker caused a significant increase in pericyte
in Tie2 activity. Studies in cultured endothelial coverage and in PECAM localization at endo-
cells have shown that Tie2 activation promotes thelial cell junctions, these effects were reversed
endothelial cell survival (Kim et al. 2000a). by the Ang1-specific blocker (Falcon et al.
Whether increased endothelial cell apoptosis 2009), suggesting that in this case the effects
also contributes to the decreased tumor vascular- of Ang2 blockade result from an increase in
ity that is observed following Ang2 blockade Tie2 activity. One could imagine that different
remains to be determined. subsets of tumor vessels are exposed to different
Although the role of Ang2 in tumors was local concentrations of Ang2/Ang1 and that
initially believed to be antagonism of Tie2, this determines the net effect of Ang2 blockade
some data support a model in which Ang2 on Tie2 phosphorylation. For example, if most
promotes tumor growth via Tie2 activation. of the Tie2 agonist activity in sprouting vessels
Support for this alternate model comes from is mediated by Ang2, then Ang2 blockade would
studies of tumors (teratomas) formed from be expected to decrease Tie2 phosphorylation in
embryonic stem cells that lack VE-PTP, a Tie2 these cells, even though Ang2 is a relatively weak
phosphatase that is specifically expressed in agonist. If, on the other hand, there are some-
the vasculature (Li et al. 2009). When implanted what higher levels of Ang1 in the vessels that

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650 9


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

G. Thurston and C. Daly

are normalized following Ang2 blockade, then and has emerged as a preferred target for thera-
Ang2 inhibition would increase Tie2 phosphor- peutic blockade. Despite our poor understand-
ylation by allowing greater access of Ang1 to ing of the precise functional role of Ang2, two
Tie2. Although further work is clearly required things are clear: Ang2 expression is very often
to substantiate the agonist role of Ang2 in increased in vascular remodeling and disease;
tumors, the available data strongly suggest that and blocking Ang2 appears to benefit the course
the effects of Ang2 blockade on tumor vascular- of the disease, whether it is airway inflamma-
ity and growth result from a decrease in Tie2 tion, lung injury, or solid tumors.
signaling. Several key aspects of Ang2 biology need
The picture that has emerged from these further investigation. One issue is to determine
preclinical studies is that Ang2 blockade inhib- whether Ang2 produced by endothelial cells and
its the growth of a broad range of tumors but acting in an autocrine manner is functionally
that the effects on growth are relatively modest. similar to recombinant exogenous Ang2 (which
This is in contrast to anti-VEGF agents, which has been applied in many experimental set-
provide complete growth inhibition in some tings). If not, then what are the biochemical
of the same preclinical tumor models. Given mechanisms? A second issue is to determine
that blockade of VEGF has proven to be benefi- what factors are responsible for the up-regula-
cial but not curative in several human cancers tion of Ang2 in disease settings. Although sev-
(Duda et al. 2007), several groups have exam- eral factors have been reported to induce Ang2
ined whether Ang2 blockade can potentiate expression in cultured cells, it is not clear if
the effects of VEGF inhibitors. Combined treat- these are the relevant factors in vivo. Another
ment with Ang2 and VEGF blockers has been key issue is to determine whether endogenous
shown to provide better inhibition of tumor Ang2 can have different actions on Tie2 in dif-
www.perspectivesinmedicine.org

growth than the single agents in a number of ferent cellular contexts. An extension of this
tumor models, including some that exhibit a issue is to determine whether Tie2 activation
very strong response to VEGF blockade (Brown (by either Ang1 or Ang2) can have different cel-
et al. 2010; Hashizume et al. 2010; Huang et al. lular effects in different contexts. To address this
2010). Consistent with the more potent effects particular issue, we need to understand more
on tumor growth, the combination treatment about the key factors within tumor and inflam-
reduces tumor vascularity to a much greater matory environments that determine the con-
extent than the single agents do (Hashizume text. For example, we need to understand more
et al. 2010). Thus, it appears that inhibition of about how angiopoietin Tie signaling interacts
Ang2 imposes an additional stress on tumor with VEGF signaling. Finally, although several
endothelial cells beyond that provided by VEGF reports have described interactions of other
blockade alone. The precise nature of the molec- proteins with angiopoietins and Tie2, it will
ular events that ensue following Ang2 versus be important to determine whether these inter-
VEGF blockade and how these events combine actions contribute functionally to the activity of
to impair endothelial cell function remain an angiopoietins in diseases settings. Led by the
exciting subject for further investigation. ongoing clinical development of Ang2 inhibi-
tors, the next few years promise to be another
exciting period for research into the angiopoie-
SUMMARY AND OUTSTANDING ISSUES
tin Tie pathway.
In recent years, the components of the angio-
poietin Tie pathway have been characterized.
ACKNOWLEDGMENTS
However, as is evident from this review, the net-
worked effects of these components on vascular We thank the many outstanding members of
biology and pathology are not fully understood. the Regeneron scientific community who have
In this review, we have focused on Ang2 because helped shape our understanding of the angio-
it has been a particularly enigmatic component poietin Tie system.

10 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

Angiopoietin Tie Signaling Pathway

REFERENCES Rafique A, et al. 2003. Angiopoietins have distinct mod-


ular domains essential for receptor binding, dimerization
Augustin HG, Koh GY, Thurston G, Alitalo K. 2009. Control and superclustering. Nat Struct Biol 10: 38 44.
of vascular morphogenesis and homeostasis through the Davis JS, Yeo TW, Piera KA, Woodberry T, Celermajer DS,
angiopoietin Tie system. Nat Rev Mol Cell Biol 10: Stephens DP, Anstey NM. 2010. Angiopoietin-2 is
165177. increased in sepsis and inversely associated with nitric
Baffert F, Thurston G, Rochon-Duck M, Le T, Brekken R, oxide-dependent microvascular reactivity. Crit Care 14:
McDonald DM. 2004. Age-related changes in vascular R89. doi: 10.1186/cc9020.
endothelial growth factor dependency and angiopoie- Detjen KM, Rieke S, Deters A, Schulz P, Rexin A, Vollmer
tin-1-induced plasticity of adult blood vessels. Circ Res S, Hauff P, Wiedenmann B, Pavel M, Scholz A. 2010.
94: 984 992. Angiopoietin-2 promotes disease progression of neuro-
Baumer S, Keller L, Holtmann A, Funke R, August B, Gamp endocrine tumors. Clin Cancer Res 16: 420 429.
A, Wolburg H, Wolburg-Buchholz K, Deutsch U, Vest- Dominguez MG, Hughes VC, Pan L, Simmons M, Daly C,
weber D. 2006. Vascular endothelial cell-specific phos- Anderson K, Noguera-Troise I, Murphy AJ, Valenzuela
photyrosine phosphatase (VE-PTP) activity is required DM, Davis S, et al. 2007. Vascular endothelial tyrosine
for blood vessel development. Blood 107: 4754 4762. phosphatase (VE-PTP)-null mice undergo vasculogene-
Bhandari V, Choo-Wing R, Lee CG, Zhu Z, Nedrelow JH, sis but die embryonically because of defects in angiogen-
Chupp GL, Zhang X, Matthay MA, Ware LB, Homer esis. Proc Natl Acad Sci 104: 3243 3248.
RJ, et al. 2006. Hyperoxia causes angiopoietin 2-mediated Duda DG, Batchelor TT, Willett CG, Jain RK. 2007. VEGF-
acute lung injury and necrotic cell death. Nat Med 12: targeted cancer therapy strategies: Current progress, hur-
1286 1293. dles and future prospects. Trends Mol Med 13: 223 230.
Brown LF, Dezube BJ, Tognazzi K, Dvorak HF, Yancopoulos Dumont DJ, Gradwohl G, Fong GH, Puri MC, Gertsenstein
GD. 2000. Expression of Tie1, Tie2, and angiopoietins 1, M, Auerbach A, Breitman ML. 1994. Dominant-negative
2, and 4 in Kaposis sarcoma and cutaneous angiosar- and targeted null mutations in the endothelial receptor
coma. Am J Pathol 156: 21792183. tyrosine kinase, tek, reveal a critical role in vasculogenesis
Brown JL, Cao ZA, Pinzon-Ortiz M, Kendrew J, Reimer C, of the embryo. Genes Dev 8: 1897 1909.
Wen S, Zhou JQ, Tabrizi M, Emery S, McDermott B, Durkin AJ, Bloomston M, Yeatman TJ, Gilbert-Barness E,
et al. 2010. A human monoclonal anti-ANG2 antibody Cojita D, Rosemurgy AS, Zervos EE. 2004. Differential
leads to broad antitumor activity in combination with expression of the Tie-2 receptor and its ligands in human
www.perspectivesinmedicine.org

VEGF inhibitors and chemotherapy agents in preclinical pancreatic tumors. J Am Coll Surg 199: 724 731.
models. Mol Cancer Ther 9: 145 156. Emoto M, Charnock-Jones DS, Licence DR, Ishiguro M,
Chen L, Yang Z, Wang G, Wang C. 2001. Expression of Kawai M, Yanaihara A, Saito T, Hachisuga T, Iwasaki H,
angiopoietin-2 gene and its receptor Tie2 in hepatocellu- Kawarabayashi T, et al. 2004. Localization of the VEGF
lar carcinoma. J Tongji Med Univ 21: 228 235. and angiopoietin genes in uterine carcinosarcoma. Gyne-
Cho CH, Kammerer RA, Lee HJ, Steinmetz MO, Ryu YS, Lee col Oncol 95: 474 482.
SH, Yasunaga K, Kim KT, Kim I, Choi HH, et al. 2004. Falcon BL, Hashizume H, Koumoutsakos P, Chou J, Bready
COMP-Ang1: A designed angiopoietin-1 variant with JV, Coxon A, Oliner JD, McDonald DM. 2009. Contrast-
nonleaky angiogenic activity. Proc Natl Acad Sci 101: ing actions of selective inhibitors of angiopoietin-1 and
5547 5552. angiopoietin-2 on the normalization of tumor blood ves-
Coxon A, Bready J, Min H, Kaufman S, Leal J, Yu D, Lee sels. Am J Pathol 175: 21592170.
TA, Sun JR, Estrada J, Bolon B, et al. 2010. Context- Fiedler U, Scharpfenecker M, Koidl S, Hegen A, Grunow V,
dependent role of angiopoietin-1 inhibition in the sup- Schmidt JM, Kriz W, Thurston G, Augustin HG. 2004.
pression of angiogenesis and tumor growth: Implications The Tie-2 ligand angiopoietin-2 is stored in and rapidly
for AMG 386, an angiopoietin-1/2-neutralizing pepti- released upon stimulation from endothelial cell Weibel-
body. Mol Cancer Ther 9: 26412651. Palade bodies. Blood 103: 41504156.
Daly C, Wong V, Burova E, Wei Y, Zabski S, Griffiths J, Lai Fiedler U, Reiss Y, Scharpfenecker M, Grunow V, Koidl S,
KM, Lin HC, Ioffe E, Yancopoulos GD, et al. 2004. Thurston G, Gale NW, Witzenrath M, Rosseau S, Suttorp
Angiopoietin-1 modulates endothelial cell function and N, et al. 2006. Angiopoietin-2 sensitizes endothelial cells
gene expression via the transcription factor FKHR to TNF-a and has a crucial role in the induction of
(FOXO1). Genes Dev 18: 1060 1071. inflammation. Nat Med 12: 235 239.
Daly C, Pasnikowski E, Burova E, Wong V, Aldrich TH, Grif- Fukuhara S, Sako K, Minami T, Noda K, Kim HZ, Kodama T,
fiths J, Ioffe E, Daly TJ, Fandl JP, Papadopoulos N, et al. Shibuya M, Takakura N, Koh GY, Mochizuki N. 2008.
2006. Angiopoietin-2 functions as an autocrine protec- Differential function of Tie2 at cell cell contacts and
tive factor in stressed endothelial cells. Proc Natl Acad cell substratum contacts regulated by angiopoietin-1.
Sci 103: 15491 15496. Nat Cell Biol 10: 513 526.
Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Fuxe J, Lashnits E, OBrien S, Baluk P, Tabruyn SP, Kuhnert F,
Jain V, Ryan TE, Bruno J, Radziejewski C, Maisonpierre Kuo C, Thurston G, McDonald DM. 2010. Angiopoie-
PC, et al. 1996. Isolation of angiopoietin-1, a ligand for tin/Tie2 signaling transforms capillaries into venules
the TIE2 receptor, by secretion-trap expression cloning. primed for leukocyte trafficking in airway inflammation.
Cell 87: 11611169. Am J Pathol 176: 2009 2018.
Davis S, Papadopoulos N, Aldrich TH, Maisonpierre PC, Gale NW, Thurston G, Hackett SF, Renard R, Wang Q,
Huang T, Kovac L, Xu A, Leidich R, Radziejewska E, McClain J, Martin C, Witte C, Witte MH, Jackson D,

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650 11


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

G. Thurston and C. Daly

et al. 2002. Angiopoietin-2 is required for postnatal VCAM-1, and E-selectin expression. Circ Res 89:
angiogenesis and lymphatic patterning, and only the 477 479.
latter role is rescued by Angiopoietin-1. Dev Cell 3: Li JJ, Huang YQ, Basch R, Karpatkin S. 2001. Thrombin
411423. induces the release of angiopoietin-1 from platelets.
Gamble JR, Drew J, Trezise L, Underwood A, Parsons M, Thromb Haemost 85: 204 206.
Kasminkas L, Rudge J, Yancopoulos G, Vadas MA. Li X, Hahn CN, Parsons M, Drew J, Vadas MA, Gamble JR.
2000. Angiopoietin-1 is an antipermeability and anti- 2004. Role of protein kinase C6 in thrombin-induced
inflammatory agent in vitro and targets cell junctions. endothelial permeability changes: Inhibition by
Circ Res 87: 603 607. angiopoietin-1. Blood 104: 17161724.
Goede V, Coutelle O, Neuneier J, Reinacher-Schick A, Li Z, Huang H, Boland P, Dominguez MG, Burfeind P, Lai
Schnell R, Koslowsky TC, Weihrauch MR, Cremer B, KM, Lin HC, Gale NW, Daly C, Auerbach W, et al.
Kashkar H, Odenthal M, et al. 2010. Identification 2009. Embryonic stem cell tumor model reveals role of
of serum angiopoietin-2 as a biomarker for clinical vascular endothelial receptor tyrosine phosphatase in
outcome of colorectal cancer patients treated with regulating Tie2 pathway in tumor angiogenesis. Proc
bevacizumab-containing therapy. Br J Cancer 103: Natl Acad Sci 106: 22399 22404.
1407 1414. Limaye N, Wouters V, Uebelhoer M, Tuominen M, Wirkkala
Hanahan D. 1997. Signaling vascular morphogenesis and R, Mulliken JB, Eklund L, Boon LM, Vikkula M. 2009.
maintenance. Science 277: 48 50. Somatic mutations in angiopoietin receptor gene TEK
Hashizume H, Falcon BL, Kuroda T, Baluk P, Coxon A, Yu D, cause solitary and multiple sporadic venous malforma-
Bready JV, Oliner JD, McDonald DM. 2010. Comple- tions. Nat Genet 41: 118 124.
mentary actions of inhibitors of angiopoietin-2 and Lin P, Buxton JA, Acheson A, Radziejewski C, Maisonpierre
VEGF on tumor angiogenesis and growth. Cancer Res PC, Yancopoulos GD, Channon KM, Hale LP, Dewhirst
70: 22132223. MW, George SE, et al. 1998. Antiangiogenic gene therapy
Helfrich I, Edler L, Sucker A, Thomas M, Christian S, Scha- targeting the endothelium-specific receptor tyrosine ki-
dendorf D, Augustin HG. 2009. Angiopoietin-2 levels are nase Tie2. Proc Natl Acad Sci 95: 8829 8834.
associated with disease progression in metastatic malig- Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand
nant melanoma. Clin Cancer Res 15: 1384 1392. SJ, Radziejewski C, Compton D, McClain J, Aldrich TH,
Papadopoulos N, et al. 1997. Angiopoietin-2, a natural
Holash J, Wiegand SJ, Yancopoulos GD. 1999. New model of
antagonist for Tie2 that disrupts in vivo angiogenesis.
www.perspectivesinmedicine.org

tumor angiogenesis: Dynamic balance between vessel


Science 277: 5560.
regression and growth mediated by angiopoietins and
VEGF. Oncogene 18: 5356 5362. Mandriota SJ, Pepper MS. 1998. Regulation of angiopoietin-
2 mRNA levels in bovine microvascular endothelial cells
Huang H, Lai JY, Do J, Liu D, Li L, Del Rosario J, Dop-
by cytokines and hypoxia. Circ Res 83: 852 859.
palapudi VR, Pirie-Shepherd S, Levin N, Bradshaw C,
et al. 2010. Specifically targeting angiopoietin-2 inhibits Moon WS, Park HS, Yu KH, Jang KY, Kang MJ, Park H, Tar-
angiogenesis, Tie2-expressing monocyte infiltration, nawski AS. 2006. Expression of angiopoietin 1, 2 and
and tumor growth. Clin Cancer Res 17: 10011011. their common receptor Tie2 in human gastric carcinoma:
implication for angiogenesis. J Korean Med Sci 21:
Jho D, Mehta D, Ahmmed G, Gao XP, Tiruppathi C, Broman 272 278.
M, Malik AB. 2005. Angiopoietin-1 opposes VEGF-
induced increase in endothelial permeability by in- Nasarre P, Thomas M, Kruse K, Helfrich I, Wolter V, Depper-
hibiting TRPC1-dependent Ca2 influx. Circ Res 96: mann C, Schadendorf D, Thurston G, Fiedler U, Augus-
tin HG. 2009. Host-derived angiopoietin-2 affects early
1282 1290.
stages of tumor development and vessel maturation but
Joussen AM, Poulaki V, Tsujikawa A, Qin W, Qaum T, Xu Q, is dispensable for later stages of tumor growth. Cancer
Moromizato Y, Bursell SE, Wiegand SJ, Rudge J, et al. Res 69: 1324 1333.
2002. Suppression of diabetic retinopathy with angio-
Nykanen AI, Pajusola K, Krebs R, Keranen MA, Raisky O,
poietin-1. Am J Pathol 160: 16831693.
Koskinen PK, Alitalo K, Lemstrom KB. 2006. Common
Kim I, Kim HG, So JN, Kim JH, Kwak HJ, Koh GY. protective and diverse smooth muscle cell effects of AAV-
2000a. Angiopoietin-1 regulates endothelial cell survival mediated angiopoietin-1 and -2 expression in rat cardiac
through the phosphatidylinositol 30 -kinase/Akt signal allograft vasculopathy. Circ Res 98: 13731380.
transduction pathway. Circ Res 86: 24 29. Oliner J, Min H, Leal J, Yu D, Rao S, You E, Tang X, Kim H,
Kim I, Kim JH, Moon SO, Kwak HJ, Kim NG, Koh GY. Meyer S, Han SJ, et al. 2004. Suppression of angiogenesis
2000b. Angiopoietin-2 at high concentration can and tumor growth by selective inhibition of angiopoie-
enhance endothelial cell survival through the phosphati- tin-2. Cancer Cell 6: 507 516.
dylinositol 30 -kinase/Akt signal transduction pathway. Puri MC, Rossant J, Alitalo K, Bernstein A, Partanen J. 1995.
Oncogene 19: 45494552. The receptor tyrosine kinase TIE is required for integrity
Kim I, Kim JH, Ryu YS, Liu M, Koh GY. 2000c. Tumor and survival of vascular endothelial cells. EMBO J 14:
necrosis factor-a upregulates angiopoietin-2 in human 58845891.
umbilical vein endothelial cells. Biochem Biophys Res Saharinen P, Kerkela K, Ekman N, Marron M, Brindle N, Lee
Commun 269: 361365. GM, Augustin H, Koh GY, Alitalo K. 2005. Multiple
Kim I, Moon SO, Park SK, Chae SW, Koh GY. 2001. angiopoietin recombinant proteins activate the Tie1
Angiopoietin-1 reduces VEGF-stimulated leukocyte receptor tyrosine kinase and promote its interaction
adhesion to endothelial cells by reducing ICAM-1, with Tie2. J Cell Biol 169: 239 243.

12 Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

Angiopoietin Tie Signaling Pathway

Saharinen P, Eklund L, Miettinen J, Wirkkala R, Anisimov A, blood vessels in mice transgenically overexpressing
Winderlich M, Nottebaum A, Vestweber D, Deutsch U, angiopoietin-1. Science 286: 25112514.
Koh GY, et al. 2008. Angiopoietins assemble distinct Thurston G, Rudge JS, Ioffe E, Zhou H, Ross L, Croll SD,
Tie2 signalling complexes in endothelial cellcell and Glazer N, Holash J, McDonald DM, Yancopoulos GD.
cell matrix contacts. Nat Cell Biol 10: 527537. 2000. Angiopoietin-1 protects the adult vasculature
Sato TN, Qin Y, Kozak CA, Audus KL. 1993. Tie-1 and tie-2 against plasma leakage. Nat Med 6: 460 463.
define another class of putative receptor tyrosine kinase
Thurston G, Wang Q, Baffert F, Rudge J, Papadopoulos N,
genes expressed in early embryonic vascular system.
Proc Natl Acad Sci 90: 93559358. Jean-Guillaume D, Wiegand S, Yancopoulos GD, McDo-
nald DM. 2005. Angiopoietin 1 causes vessel enlarge-
Sato TN, Tozawa Y, Deutsch U, Wolburg-Buchholz K, Fuji-
ment, without angiogenic sprouting, during a critical
wara Y, Gendron-Maguire M, Gridley T, Wolburg H,
developmental period. Development 132: 3317 3326.
Risau W, Qin Y. 1995. Distinct roles of the receptor tyro-
sine kinases Tie-1 and Tie-2 in blood vessel formation. van der Heijden M, van Nieuw Amerongen GP, Chedamni S,
Nature 376: 7074. van Hinsbergh VW, Johan Groeneveld AB. 2009. The
Scholz A, Rehm VA, Rieke S, Derkow K, Schulz P, Neumann angiopoietin Tie2 system as a therapeutic target in sep-
K, Koch I, Pascu M, Wiedenmann B, Berg T, et al. 2007. sis and acute lung injury. Expert Opin Ther Targets 13:
Angiopoietin-2 serum levels are elevated in patients 39 53.
with liver cirrhosis and hepatocellular carcinoma. Am J Vikkula M, Boon LM, Carraway KL III, Calvert JT, Diamonti
Gastroenterol 102: 24712481. AJ, Goumnerov B, Pasyk KA, Marchuk DA, Warman ML,
Stratmann A, Risau W, Plate KH. 1998. Cell type-specific Cantley LC, et al. 1996. Vascular dysmorphogenesis
expression of angiopoietin-1 and angiopoietin-2 suggests caused by an activating mutation in the receptor tyrosine
a role in glioblastoma angiogenesis. Am J Pathol 153: kinase TIE2. Cell 87: 1181 1190.
1459 1466. Winderlich M, Keller L, Cagna G, Broermann A, Kamenyeva
Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, O, Kiefer F, Deutsch U, Nottebaum AF, Vestweber D.
Davis S, Sato TN, Yancopoulos GD. 1996. Requisite role 2009. VE-PTP controls blood vessel development by bal-
of angiopoietin-1, a ligand for the TIE2 receptor, during ancing Tie-2 activity. J Cell Biol 185: 657 671.
embryonic angiogenesis. Cell 87: 11711180.
Witzenbichler B, Westermann D, Knueppel S, Schultheiss
Suri C, McClain J, Thurston G, McDonald DM, Zhou H, HP, Tschope C. 2005. Protective role of angiopoietin-1
Oldmixon EH, Sato TN, Yancopoulos GD. 1998. In-
www.perspectivesinmedicine.org

in endotoxic shock. Circulation 111: 97 105.


creased vascularization in mice overexpressing angio-
poietin-1. Science 282: 468 471. Xu Y, Yu Q. 2001. Angiopoietin-1, unlike angiopoietin-2, is
incorporated into the extracellular matrix via its linker
Tabruyn SP, Colton K, Morisada T, Fuxe J, Wiegand SJ,
peptide region. J Biol Chem 276: 34990 34998.
Thurston G, Coyle AJ, Connor J, McDonald DM. 2010.
Angiopoietin-2-driven vascular remodeling in airway Yuan HT, Venkatesha S, Chan B, Deutsch U, Mammoto T,
inflammation. Am J Pathol 177: 32333243. Sukhatme VP, Woolf AS, Karumanchi SA. 2007. Activa-
Teichert-Kuliszewska K, Maisonpierre PC, Jones N, Camp- tion of the orphan endothelial receptor Tie1 modifies
bell AI, Master Z, Bendeck MP, Alitalo K, Dumont DJ, Tie2-mediated intracellular signaling and cell survival.
Yancopoulos GD, Stewart DJ. 2001. Biological action of FASEB J 21: 31713183.
angiopoietin-2 in a fibrin matrix model of angiogenesis Zagzag D, Hooper A, Friedlander DR, Chan W, Holash J,
is associated with activation of Tie2. Cardiovasc Res 49: Wiegand SJ, Yancopoulos GD, Grumet M. 1999. In situ
659670. expression of angiopoietins in astrocytomas identifies
Thurston G, Suri C, Smith K, McClain J, Sato TN, Yanco- angiopoietin-2 as an early marker of tumor angiogenesis.
poulos GD, McDonald DM. 1999. Leakage-resistant Exp Neurol 159: 391 400.

Cite this article as Cold Spring Harb Perspect Med 2012;2:a006650 13


Downloaded from http://perspectivesinmedicine.cshlp.org/ on April 13, 2015 - Published by Cold Spring Harbor Laboratory Press

The Complex Role of Angiopoietin-2 in the AngiopoietinTie Signaling


Pathway
Gavin Thurston and Christopher Daly

Cold Spring Harb Perspect Med 2012; doi: 10.1101/cshperspect.a006650 originally published online April
10, 2012

Subject Collection Angiogenesis

miRNAs as Modulators of Angiogenesis Arteriovenous Malformations and Other Vascular


Shira Landskroner-Eiger, Isabelle Moneke and Malformation Syndromes
William C. Sessa Kevin J. Whitehead, Matthew C.P. Smith and Dean
Y. Li
VEGF and Notch in Tip and Stalk Cell Selection Molecular Parallels between Neural and Vascular
Raquel Blanco and Holger Gerhardt Development
Anne Eichmann and Jean-Lon Thomas
The Role of the Tumor Microenvironment in The VEGF Pathway in Cancer and Disease:
Regulating Angiogenesis Responses, Resistance, and the Path Forward
Randolph S. Watnick Mark W. Kieran, Raghu Kalluri and Yoon-Jae Cho
Angiogenic Factors in Preeclampsia and Related Common Polymorphisms in Angiogenesis
Disorders Michael S. Rogers and Robert J. D'Amato
Ana Sofia Cerdeira and S. Ananth Karumanchi
Anti-VEGF Therapies in the Clinic Endothelial Cell-to-Cell Junctions: Adhesion and
Kellen L. Meadows and Herbert I. Hurwitz Signaling in Physiology and Pathology
Maria Grazia Lampugnani
The Complex Role of Angiopoietin-2 in the VEGF-Directed Blood Vessel Patterning: From
AngiopoietinTie Signaling Pathway Cells to Organism
Gavin Thurston and Christopher Daly Victoria L. Bautch
PlGF: A Multitasking Cytokine with Vascular Anomalies: From Genetics toward
Disease-Restricted Activity Models for Therapeutic Trials
Mieke Dewerchin and Peter Carmeliet Melanie Uebelhoer, Laurence M. Boon and Miikka
Vikkula
Human Endothelial Progenitor Cells Signal Transduction by Vascular Endothelial
Mervin C. Yoder Growth Factor Receptors
Sina Koch and Lena Claesson-Welsh

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright 2012 Cold Spring Harbor Laboratory Press; all rights reserved

Вам также может понравиться