ArticlePDF AvailableLiterature Review

Taming of the wild vessel: Promoting vessel stabilization for safe therapeutic angiogenesis

Authors:

Abstract and Figures

VEGF (vascular endothelial growth factor) is the master regulator of blood vessel growth. However, it displayed substantial limitations when delivered as a single gene to restore blood flow in ischaemic conditions. Indeed, uncontrolled VEGF expression can easily induce aberrant vascular structures, and short-term expression leads to unstable vessels. Targeting the second stage of the angiogenic process, i.e. vascular maturation, is an attractive strategy to induce stable and functional vessels for therapeutic angiogenesis. The present review discusses the limitations of VEGF-based gene therapy, briefly summarizes the current knowledge of the molecular and cellular regulation of vascular maturation, and describes recent pre-clinical evidence on how the maturation stage could be targeted to achieve therapeutic angiogenesis.
Content may be subject to copyright.
1654 Biochemical Society Transactions (2011) Volume 39, part 6
Taming of the wild vessel: promoting vessel
stabilization for safe therapeutic angiogenesis
Silvia Reginato, Roberto Gianni-Barrera and Andrea Banfi1
Cell and Gene Therapy, Department of Biomedicine and Department of Surgery, Basel University Hospital, Hebelstrasse 20, Basel, CH-4031, Switzerland
Abstract
VEGF (vascular endothelial growth factor) is the master regulator of blood vessel growth. However, it
displayed substantial limitations when delivered as a single gene to restore blood flow in ischaemic
conditions. Indeed, uncontrolled VEGF expression can easily induce aberrant vascular structures, and short-
term expression leads to unstable vessels. Targeting the second stage of the angiogenic process, i.e. vascular
maturation, is an attractive strategy to induce stable and functional vessels for therapeutic angiogenesis.
The present review discusses the limitations of VEGF-based gene therapy, briefly summarizes the current
knowledge of the molecular and cellular regulation of vascular maturation, and describes recent pre-clinical
evidence on how the maturation stage could be targeted to achieve therapeutic angiogenesis.
Introduction
The growth of new blood vessels (angiogenesis) is a
fundamental process to allow tissue repair and regeneration
and plays also a crucial role in many pathological conditions,
such as cancer, inflammation and ischaemia. In particular,
therapeutic angiogenesis, i.e. the stimulation of vascular
growth by the delivery of specific growth factors, is
an attractive strategy to restore blood flow in chronic-
ally ischaemic tissue. VEGF (vascular endothelial growth
factor) is the master regulator of angiogenesis in both
development and postnatal life. However, angiogenesis is
a complex multi-step process that requires the temporal
and spatial co-ordination of different factors and cell types.
Endothelial cells proliferate and migrate in response to VEGF
gradients to form new tubular structures. Subsequently, the
maturation of new blood vessels requires the recruitment
of mural cells (pericytes or smooth muscle cells) and the
deposition of a basal lamina [1]. The maturation stage is
crucial to ensure the return to quiescence of the activated
endothelium, as well as the persistence and the functionality
of the new vascular structures. Therefore an understanding
of the cellular and molecular cross-talk during the different
stages of vascular growth is crucial to design therapeutic
strategies that are both safe and effective.
Limitations of VEGF gene delivery for
therapeutic angiogenesis
The results of placebo-controlled clinical trials of VEGF gene
therapy for both peripheral and coronary artery disease were
Key words: angiopoietin, neuropilin-1, platelet-derived growth factor BB (PDGF-BB),
transforming growth factor β(TGFβ), vascular endothelial growth factor (VEGF), vascular
maturation.
Abbreviations used: AAV, adeno-associated vector; Ang1/2, angiopoietin 1/2; CXCR4, CXC
chemokine receptor 4; NEM cell, neuropilin-1-expressing mononuclear cell; NRP1, neuropilin-1;
PDGF-BB, platelet-derived growth factor BB; TGFβ, transforming growth factor β;VEGF,vascular
endothelial growth factor.
1To whom correspondence should be addressed (email abanfi@uhbs.ch).
disappointing [2], and, although VEGF gene delivery was
safe, it did not generate sufficient angiogenesis to correct the
underlying ischaemia. Retrospective analyses have uncovered
several factors that had not been adequately taken into
consideration in designing the first clinical trials and that
could account for the therapeutic results [3]. Of these, we
focus on two limitations of VEGF-based gene therapy that
arise from biological properties of VEGF itself, namely
the need to control its dose distribution in vivo and the
requirement for prolonged expression.
Several lines of evidence suggest that VEGF gene delivery
has a very narrow therapeutic window in vivo: low doses have
no or little angiogenic effects, whereas higher doses rapidly
become unsafe, inducing vessels that frequently display mor-
phological and functional abnormalities. Exogenous VEGF
administration during embryonic vasculogenesis results in
profoundly altered development of vessels with large lumens
[4]. Furthermore, transgenic mice overexpressing VEGF in
the skin, as well as in the heart and liver, display malformed
leaky vessels with unusually large and irregular lumens
[5,6]. The induction of vascular tumours (haemangiomas)
as a consequence of uncontrolled VEGF expression was
also reported in skeletal muscle [7] and in the myocardium
[8] after the delivery of retrovirally transduced myoblasts.
Adenoviral overexpression of VEGF in the skin, fat, heart
and skeletal muscle of mice caused the formation of enlarged,
thin-walled, pericyte-poor vessels, as well as multi-lumenized
glomeruloid structures that resembled tumour-associated
vascular malformations [9,10]. Finally, angioma growth was
observed in infarcted rat hearts after the injection of a VEGF-
encoding plasmid [11].
Previous work from our group has carefully investigated
the dose-dependent effects of VEGF in both normal and
ischaemic skeletal muscle [12,13]. These results indicate that
VEGF does not have an intrinsically narrow dose–response
curve. However, VEGF induces normal or aberrant an-
giogenesis depending on its amount in the microenvironment
C
The Authors Journal compilation C
2011 Biochemical Society Biochem. Soc. Trans. (2011) 39, 1654–1658; doi:10.1042/BST20110652
Biochemical Society Transactions www.biochemsoctrans.org
Advances in the Cellular and Molecular Biology of Angiogenesis 1655
around each producing cell, and not on its total dose, as it
remains localized in the extracellular matrix [14]. Therefore
direct in vivo gene therapy approaches, which generate
heterogeneous expression levels around each transduced cell,
lead to a waste of the therapeutic window [14].
Duration of expression is another crucial point to
be considered when delivering VEGF for therapeutic
angiogenesis. On one hand, it is desirable to avoid long-
term expression of a potent growth factor such as VEGF,
but on the other, too brief expression is ineffective. In
a transgenic system of conditional switching of VEGF
expression, it has been shown that VEGF production
for approximately 2 weeks leads to unstable vessels that
promptly regress as soon as the angiogenic stimulus is
stopped. However, if expression was prolonged for at least
4 weeks, the new vessels matured and persisted indefinitely
after VEGF withdrawal [5]. VEGF-dependence of newly
induced vessels has been demonstrated also by injecting
inducible AAVs (adeno-associated vectors) in skeletal muscle,
confirming that sustained VEGF expression for at least 1
month is needed to induce the formation of stable vessels
[15]. Furthermore, abrogation of VEGF signalling with
a recombinant receptor-body (VEGF-Trap) did not cause
regression of new vessels only 4 weeks after implantation of
VEGF-expressing myoblasts in skeletal muscle. Interestingly,
aberrant vessels induced by high VEGF levels never became
VEGF-independent [12].
The need for at least 4 weeks of sustained VEGF expression
in order to generate stable vessels is a challenge for the use
of short-term gene-delivery systems, such as plasmids and
adenoviral vectors, for therapeutic angiogenesis.
Mechanisms of vascular maturation
Vascular maturation, which involves the induction of
endothelial quiescence and protection against VEGF with-
drawal, requires a tight co-ordination of different cell types,
including endothelial cells, smooth muscle cells/pericytes
and recently described populations of myeloid cells [16]
(Figure 1).
Nascent endothelial tubes are coated by pericytes.
During vessel sprouting, pericytes are recruited by PDGF-
BB (platelet-derived growth factor BB) produced by the
migrating tip cell. Genetic studies revealed that mice lacking
PDGF-BB or PDGFRβ(platelet-derived growth factor
receptor β) genes display vascular abnormalities, formation
of microaneurysms and bleeding [17,18]. Pericyte deficiency
in mice lacking the NG2 (nerve/glial antigen-2) proteoglycan
results in immature tumour vessels and impaired basal lamina
assembly [19].
The association between endothelium and pericytes
renders new vessels independent of continued VEGF expres-
sion, as demonstrated in the vascularization of the neonatal
retina as well as in tumours [20,21]. In diabetic retinopathy,
excessive VEGF production leads to the formation of
pericyte-poor and leaky vessels, resulting in blindness [22].
Continuous uncontrolled VEGF overexpression in skeletal
Figure 1 Cellular and molecular cross-talk during vascular
maturation
(A) VEGF drives the release of Ang2, which inhibits Tie2 signalling and
causes the activation of quiescent endothelial cells. Tip cells (yellow)
initiate sprouting and produce PDGF-BB. Trailing endothelial cells (green)
proliferate to form the stalk of the new vessels, which, at this stage, are
not yet invested by pericytes (orange). (B) During vascular maturation,
pericytes, recruited by PDGF-BB, make contact with the new endothelial
structures and exchange signals through different pathways, foremost
among which are the Ang1/Tie2 and TGFβ1/TGFβR1 (TGFβreceptor 1)
axes. VEGF also induces the recruitment of CXCR4+myeloid cells (pink)
that exert a pro-angiogenic activity through paracrine signals. Another
specific subset of myeloid cells (CD11b +/Nrp-1 +, blue) express factors
directly responsible for vascular maturation.
muscle, by retrovirally transduced myoblasts, also induced
aberrant vessels that were not covered by pericytes and failed
to stabilize, remaining dependent on VEGF signalling for
survival [12].
Pericyte–endothelium cross-talk
Pericytes exert their regulatory function on endothelial
cells through both cell–cell contact and secreted signals.
In particular, the role of the TGFβ(transforming growth
factor β) and angiopoietin signalling pathways are the best
understood.
TGFβ1 regulates endothelial cell proliferation and differ-
entiation through the ALK1 and ALK5 (activin receptor-like
kinase 1 and 5) receptors [23]. Both endothelial cells and
pericytes produce TGFβ1 in an inactive form. Only when
cell–cell contact is established does the protein undergo the
cleavage of the latency-associated peptide by plasmin and
become activated [24]. TGFβ1 promotes vessel stabilization
by inhibiting the proliferation and migration of endothelial
cells and by stimulating mural cell differentiation [25].
Furthermore, it has a direct stimulatory effect on the synthesis
and deposition of extracellular matrix components [23].
Angiopoietins are the ligands for the endothelium-specific
tyrosine kinase receptor Tie2 and exert fundamental functions
in both the induction and the maturation stages of vascular
growth. Ang1 (angiopoietin 1) is expressed by pericytes and
C
The Authors Journal compilation C
2011 Biochemical Society
1656 Biochemical Society Transactions (2011) Volume 39, part 6
has been shown to inhibit vascular permeability in the skin, in
tumours and in an in vitro model of the blood–brain barrier by
acting as an agonist for the Tie2 receptor [23]. Furthermore,
Ang1 acts as a survival signal for endothelial cells [26] and
also promotes vascular stabilization by further facilitating
pericyte recruitment [27].
Ang2 (angiopoietin 2) is secreted mainly by endothelial
cells at sites of active angiogenesis and is a partial agonist of
the Tie2 receptor, which makes its actions context-dependent.
In the presence of VEGF, Ang2 promotes sprouting of new
blood vessels and remodelling of the vasculature, since it
induces the dissociation of pericytes from endothelial cells.
In the quiescent vasculature, where Tie2 is constitutively
activated by a basal Ang1 expression, Ang2 acts as a functional
antagonist of Ang1, binding preferentially to Tie2 without
inducing signal transduction and therefore destabilizing
mature vessels [28]. The vessel-destabilizing effect of Ang2
has been also demonstrated after transgenic overexpression
of Ang2 in a normal retina [29].
Myeloid accessory cells
Previous studies have revealed an intriguing role for recruited
bone-marrow-derived circulating cells in promoting both
endothelial sprouting and the stabilization of newly formed
vessels [30] (Figure 1B). In sites of active angiogenesis, VEGF
expression drives the recruitment of a population of bone-
marrow-derived CXCR4 (CXC chemokine receptor 4)-
positive myeloid cells, which are not incorporated into newly
formed vessels, but rather acquire a perivascular position
and are retained through SDF-1 (stromal-cell-derived factor
1)/CXCR4 signalling. These cells synergistically contribute
to the formation of new blood vessels through the secretion
of pro-angiogenic factors and the prevention of their
recruitment significantly inhibits neovascularization [31,32].
Further investigations demonstrated a role for another
population of bone-marrow-recruited cells in the process of
vascular maturation. Zacchigna et al. [33] demonstrated that
cells infiltrating the site of VEGF165-induced angiogenesis
in skeletal muscle are mainly CD11b+cells co-expressing
the VEGF and semaphorin-3A receptor NRP1 (neuropilin-
1), and named them NEM (NRP1-expressing mononuclear)
cells. NEM cells were shown to promote vessel maturation
through the secretion of different paracrine factors, notably
Ang1, TGFβand PBGF-BB [33]. Therefore the authors
proposed a model in which mature vessel formation relies on
the occurrence of two events: the activation of endothelial
cells, through the canonical VEGF pathway, and the
recruitment of bone-marrow-derived cells through the NRP1
receptor. These cells in turn facilitate pericyte or SMC
(smooth muscle cell) recruitment to the new vessels by
PDGF-BB production [33].
Vascular maturation as a therapeutic
target
Targeting vascular maturation is an attractive strategy to
overcome VEGF limitations that became evident in the
first generation of clinical trials. The complexity and
heterogeneity of factors involved in vascular maturation,
described above, suggest different strategies to target this
process. To date, investigations have addressed approaches
acting either directly on endothelial cell receptors, for
example through the co-delivery of Ang1, or indirectly
through the recruitment of pericytes by PDGF-BB co-
delivery.
Systemic administration of Ang1 through intravenous
adenoviral delivery has been shown to protect adult
vasculature from the lethal vascular leakage induced by
systemic VEGF expression [34]. Local co-expression of
VEGF and Ang1 has been described to reduce VEGF-
induced leakage also in rat hindlimb ischaemia [35] and
in normal rat muscle 3 months after AAV injection [36].
Moreover, adenoviral delivery of VEGF and Ang1 induced
vessels that were more mature and persisted after 4 weeks,
whereas the effect induced by VEGF alone disappeared at
the same time point [37]. Although these results clearly
demonstrate the role of Ang1 in preventing VEGF-associated
oedema, whether Ang1 co-expression might correct the
development of aberrant angioma-like structures caused by
uncontrolled VEGF levels has not been investigated.
The approach of co-delivering PDGF-BB is based on the
rationale that increasing the recruitment of pericytes could
provide a comprehensive stimulation of several pathways
in a co-ordinated fashion, thereby better reflecting the
physiological process. Delivery of recombinant VEGF and
PDGF-BB proteins from a polymeric biomaterial has been
shown to induce larger and more mature vessels compared
with VEGF alone subcutaneously and in ischaemic skeletal
muscle [38]. The combination of recombinant PDGF-BB
with FGF-2 (fibroblast growth factor-2) has been reported
to increase vascular stabilization in the mouse cornea and
in rat and rabbit hindlimb ischaemia [39]. However, the
combination of PDGF-BB was found not to correct VEGF-
induced abnormalities in the cornea and was therefore not
pursued further in the more clinically relevant muscle tissue.
Co-delivery of the VEGF and PDGF-BB genes to
ischaemic muscle and myocardium by venous retroinfusion
of AAV vectors has recently been found to significantly
improve the effects of a low VEGF vector dose, in
terms of both perfusion and collateral arteriogenesis [40].
Korpisalo et al. [41] studied the effects of VEGF and
PDGF-BB co-expression in skeletal muscle by co-delivering
two separate adenoviral vectors. They found that the
combination of VEGF and PDGF-BB did not significantly
increase angiogenesis and failed to reduce VEGF-induced
acute oedema 6 days after delivery. In addition, vascular
structures induced by the combination of the two viruses
showed an impaired mural cell coverage, since pericytes
were recruited away from the vascular structures, probably
due to the generation of discordant gradients from the
two separate vectors. Nevertheless, vessels induced by co-
expression persisted longer than those induced by VEGF
alone, through pericyte-independent paracrine signals from
recruited myeloid cells [41]. These results underline the
C
The Authors Journal compilation C
2011 Biochemical Society
Advances in the Cellular and Molecular Biology of Angiogenesis 1657
importance of a proper co-localization of VEGF and
PDGF-BB gradients in target tissue in order to stimulate
physiological angiogenesis. In fact, using myoblast-mediated
gene transfer, we recently found that only VEGF and
PDGF-BB co-expression from a single retroviral bicistronic
vector, which ensures the generation of coincidential factor
gradients around each producing cell in vivo, completely
prevented aberrant angiogenesis induced by uncontrolled
VEGF levels and induced homogeneous mature capillary
networks instead (A. Banfi, G. von Degenfeld, R. Gianni-
Barrera, M.J. Merchant, D.M. McDonald and H.M. Blau,
unpublished work). Furthermore, in the same system, we
also recently found that PDGF-BB co-expression accelerated
stabilization of vessels induced by heterogeneous VEGF
levels, so that 50% of new vessels were already VEGF-
independent after 2 weeks, whereas none was stable when
VEGF was expressed alone (S. Reginato, E. Groppa,
R. Gianni-Barrera, M. Herberer and A. Banfi, unpublished
work).
Conclusions
The increasing understanding of the biological mechanisms
that regulate blood vessels growth and maturation highlights
the complexity of molecular and cellular interactions taking
place within the angiogenic microenvironment. Therefore
targeting both vascular induction and maturation could
provide a promising strategy to overcome some limitations
related to the use of VEGF as a single factor in gene-delivery
approaches for therapeutic angiogenesis.
However, the heterogeneity of delivery systems, of
functional readouts and of target tissues considered so far in
promoting vascular maturation, generated sometimes unclear
or contradictory results. A systematic determination of both
efficacy and safety of specific combinations of factors in
clinically relevant target tissues, such as skeletal muscle for
the treatment of peripheral artery diseases, would greatly
benefit the rational design of novel therapeutic angiogenesis
strategies.
Funding
This work was supported by the Swiss National Science Foundation
[grant number 310030_127426 (to A.B.)].
References
1 Carmeliet, P. and Jain, R.K. (2011) Molecular mechanisms and clinical
applications of angiogenesis. Nature 473, 298–307
2 Gupta, R., Tongers, J. and Losordo, D.W. (2009) Human studies of
angiogenic gene therapy. Circ. Res. 105, 724–736
3 Korpisalo, P. and Yl ¨a-Herttuala, S. (2010) Stimulation of functional vessel
growth by gene therapy. Integr. Biol. 2, 102–112
4 Drake, C.J. and Little, C.D. (1995) Exogenous vascular endothelial growth
factor induces malformed and hyperfused vessels during embryonic
neovascularization. Proc. Natl. Acad. Sci. U.S.A. 92, 7657–7661
5 Dor, Y., Djonov, V., Abramovitch, R., Itin, A., Fishman, G.I., Carmeliet, P.,
Goelman, G. and Keshet, E. (2002) Conditional switching of VEGF
provides new insights into adult neovascularization and pro-angiogenic
therapy. EMBO J. 21, 1939–1947
6 Thurston, G., Suri, C., Smith, K., McClain, J., Sato, T.N., Yancopoulos, G.D.
and McDonald, D.M. (1999) Leakage-resistant blood vessels in mice
transgenically overexpressing angiopoietin-1. Science 286, 2511–2514
7 Springer, M.L., Chen, A.S., Kraft, P.E., Bednarski, M. and Blau, H.M. (1998)
VEGF gene delivery to muscle: potential role for vasculogenesis in adults.
Mol. Cell 2, 549–558
8 Lee, R.J., Springer, M.L., Blanco-Bose, W.E., Shaw, R., Ursell, P.C. and
Blau, H.M. (2000) VEGF gene delivery to myocardium: deleterious
effects of unregulated expression. Circulation 102, 898–901
9 Pettersson, A., Nagy, J.A., Brown, L.F., Sundberg, C., Morgan, E., Jungles,
S., Carter, R., Krieger, J.E., Manseau, E.J., Harvey, V.S. et al. (2000)
Heterogeneity of the angiogenic response induced in different normal
adult tissues by vascular permeability factor/vascular endothelial growth
factor. Lab. Invest. 80, 99–115
10 Sundberg, C., Nagy, J.A., Brown, L.F., Feng, D., Eckelhoefer, I.A.,
Manseau, E.J., Dvorak, A.M. and Dvorak, H.F. (2001) Glomeruloid
microvascular proliferation follows adenoviral vascular permeability
factor/vascular endothelial growth factor-164 gene delivery. Am. J.
Pathol. 158, 1145–1160
11 Schwarz, E.R., Speakman, M.T., Patterson, M., Hale, S.S., Isner, J.M.,
Kedes, L.H. and Kloner, R.A. (2000) Evaluation of the effects of
intramyocardial injection of DNA expressing vascular endothelial growth
factor (VEGF) in a myocardial infarction model in the rat: angiogenesis
and angioma formation. J. Am. Coll. Cardiol. 35, 1323–1330
12 Ozawa, C.R., Banfi, A., Glazer, N.L., Thurston, G., Springer, M.L., Kraft, P.E.,
McDonald, D.M. and Blau, H.M. (2004) Microenvironmental VEGF
concentration, not total dose, determines a threshold between normal
and aberrant angiogenesis. J. Clin. Invest. 113, 516–527
13 von Degenfeld, G., Banfi, A., Springer, M.L., Wagner, R.A., Jacobi, J.,
Ozawa, C.R., Merchant, M.J., Cooke, J.P. and Blau, H.M. (2006)
Microenvironmental VEGF distribution is critical for stable and functional
vessel growth in ischemia. FASEB J. 20, 2657–2659
14 Banfi, A., von Degenfeld, G. and Blau, H.M. (2005) Critical role of
microenvironmental factors in angiogenesis. Curr. Atheroscler. Rep. 7,
227–234
15 Tafuro, S., Ayuso, E., Zacchigna, S., Zentilin, L., Moimas, S., Dore, F. and
Giacca, M. (2009) Inducible adeno-associated virus vectors promote
functional angiogenesis in adult organisms via regulated vascular
endothelial growth factor expression. Cardiovasc. Res. 83, 663–671
16 Adams, R.H. and Alitalo, K. (2007) Molecular regulation of angiogenesis
and lymphangiogenesis. Nat. Rev. Mol. Cell Biol. 8, 464–478
17 Hellstrom, M., Kalen, M., Lindahl, P., Abramsson, A. and Betsholtz, C.
(1999) Role of PDGF-B and PDGFR-βin recruitment of vascular smooth
muscle cells and pericytes during embryonic blood vessel formation in
the mouse. Development 126, 3047–3055
18 Lindahl, P., Johansson, B.R., Leveen, P. and Betsholtz, C. (1997) Pericyte
loss and microaneurysm formation in PDGF-B-deficient mice. Science
277, 242–245
19 Huang, F.J., You, W.K., Bonaldo, P., Seyfried, T.N., Pasquale, E.B. and
Stallcup, W.B. (2010) Pericyte deficiencies lead to aberrant tumor
vascularizaton in the brain of the NG2 null mouse. Dev. Biol. 344,
1035–1046
20 Benjamin, L.E., Hemo, I. and Keshet, E. (1998) A plasticity window for
blood vessel remodelling is defined by pericyte coverage of the
preformed endothelial network and is regulated by PDGF-B and VEGF.
Development 125, 1591–1598
21 Benjamin, L.E., Golijanin, D., Itin, A., Pode, D. and Keshet, E. (1999)
Selective ablation of immature blood vessels in established human
tumors follows vascular endothelial growth factor withdrawal. J. Clin.
Invest. 103, 159–165
22 Hammes, H.P., Lin, J., Renner, O., Shani, M., Lundqvist, A., Betsholtz, C.,
Brownlee, M. and Deutsch, U. (2002) Pericytes and the pathogenesis of
diabetic retinopathy. Diabetes 51, 3107–3112
23 von Tell, D., Armulik, A. and Betsholtz, C. (2006) Pericytes and vascular
stability. Exp. Cell Res. 312, 623–629
24 Hirschi, K.K., Burt, J.M., Hirschi, K.D. and Dai, C. (2003) Gap junction
communication mediates transforming growth factor-βactivation and
endothelial-induced mural cell differentiation. Circ. Res. 93,
429–437
25 Darland, D.C. and D’Amore, P.A. (2001) TGFβis required for the
formation of capillary-like structures in three-dimensional cocultures of
10T1/2 and endothelial cells. Angiogenesis 4, 11–20
C
The Authors Journal compilation C
2011 Biochemical Society
1658 Biochemical Society Transactions (2011) Volume 39, part 6
26 Kim, I., Kim, H.G., So, J.N., Kim, J.H., Kwak, H.J. and Koh, G.Y. (2000)
Angiopoietin-1 regulates endothelial cell survival through the
phosphatidylinositol 3-kinase/Akt signal transduction pathway. Circ.
Res. 86, 24–29
27 Hawighorst, T., Skobe, M., Streit, M., Hong, Y.K., Velasco, P., Brown, L.F.,
Riccardi, L., Lange-Asschenfeldt, B. and Detmar, M. (2002) Activation of
the tie2 receptor by angiopoietin-1 enhances tumor vessel maturation
and impairs squamous cell carcinoma growth. Am. J. Pathol. 160,
1381–1392
28 Maisonpierre, P.C., Suri, C., Jones, P.F., Bartunkova, S., Wiegand, S.J.,
Radziejewski, C., Compton, D., McClain, J., Aldrich, T.H., Papadopoulos, N.
et al. (1997) Angiopoietin-2, a natural antagonist for Tie2 that disrupts
in vivo angiogenesis. Science. 277, 55–60
29 Hammes, H.P., Lin, J., Wagner, P., Feng, Y., Vom Hagen, F., Krzizok, T.,
Renner, O., Breier, G., Brownlee, M. and Deutsch, U. (2004)
Angiopoietin-2 causes pericyte dropout in the normal retina:
evidence for involvement in diabetic retinopathy. Diabetes 53,
1104–1110
30 Murdoch, C., Muthana, M., Coffelt, S.B. and Lewis, C.E. (2008) The role of
myeloid cells in the promotion of tumour angiogenesis. Nat. Rev. Cancer
8, 618–631
31 Grunewald, M., Avraham, I., Dor, Y., Bachar-Lustig, E., Itin, A., Jung, S.,
Chimenti, S., Landsman, L., Abramovitch, R. and Keshet, E. (2006)
VEGF-induced adult neovascularization: recruitment, retention, and role
of accessory cells. Cell 124, 175–189
32 Zentilin, L., Tafuro, S., Zacchigna, S., Arsic, N., Pattarini, L., Sinigaglia, M.
and Giacca, M. (2006) Bone marrow mononuclear cells are recruited to
the sites of VEGF-induced neovascularization but are not incorporated
into the newly formed vessels. Blood 107, 3546–3554
33 Zacchigna, S., Pattarini, L., Zentilin, L., Moimas, S., Carrer, A., Sinigaglia,
M., Arsic, N., Tafuro, S., Sinagra, G. and Giacca, M. (2008) Bone marrow
cells recruited through the neuropilin-1 receptor promote arterial
formation at the sites of adult neoangiogenesis in mice. J. Clin. Invest.
118, 2062–2075
34 Thurston, G., Rudge, J.S., Ioffe, E., Zhou, H., Ross, L., Croll, S.D., Glazer, N.,
Holash, J., McDonald, D.M. and Yancopoulos, G.D. (2000) Angiopoietin-1
protects the adult vasculature against plasma leakage. Nat. Med. 6,
460–463
35 Jiang, J., Jiangl, N., Gao, W., Zhu, J., Guo, Y., Shen, D., Chen, G. and Tang, J.
(2006) Augmentation of revascularization and prevention of plasma
leakage by angiopoietin-1 and vascular endothelial growth factor
co-transfection in rats with experimental limb ischaemia. Acta Cardiol.
61, 145–153
36 Arsic, N., Zentilin, L., Zacchigna, S., Santoro, D., Stanta, G., Salvi, A.,
Sinagra, G. and Giacca, M. (2003) Induction of functional
neovascularization by combined VEGF and angiopoietin-1 gene transfer
using AAV vectors. Mol. Ther. 7, 450–459
37 Zhou, Y.F., Stabile, E., Walker, J., Shou, M., Baffour, R., Yu, Z., Rott, D.,
Yancopoulos, G.D., Rudge, J.S. and Epstein, S.E. (2004) Effects of gene
delivery on collateral development in chronic hypoperfusion: diverse
effects of angiopoietin-1 versus vascular endothelial growth factor.
J. Am. Coll. Cardiol. 44, 897–903
38 Richardson, T.P., Peters, M.C., Ennett, A.B. and Mooney, D.J. (2001)
Polymeric system for dual growth factor delivery. Nat. Biotechnol. 19,
1029–1034
39 Cao, R., Brakenhielm, E., Pawliuk, R., Wariaro, D., Post, M.J., Wahlberg, E.,
Leboulch, P. and Cao, Y. (2003) Angiogenic synergism, vascular stability
and improvement of hind-limb ischemia by a combination of PDGF-BB
and FGF-2. Nat. Med. 9, 604–613
40 Kupatt, C., Hinkel, R., Pfosser, A., El-Aouni, C., Wuchrer, A., Fritz, A.,
Globisch, F., Thormann, M., Horstkotte, J., Lebherz, C. et al. (2010)
Cotransfection of vascular endothelial growth factor-A and
platelet-derived growth factor-B via recombinant adeno-associated virus
resolves chronic ischemic malperfusion role of vessel maturation. J. Am.
Coll. Cardiol. 56, 414–422
41 Korpisalo, P., Karvinen, H., Rissanen, T.T., Kilpijoki, J., Marjomaki, V.,
Baluk, P., McDonald, D.M., Cao, Y., Eriksson, U., Alitalo, K. and
Yl ¨a-Herttuala, S. (2008) Vascular endothelial growth factor-A and
platelet-derived growth factor-B combination gene therapy prolongs
angiogenic effects via recruitment of interstitial mononuclear cells and
paracrine effects rather than improved pericyte coverage of angiogenic
vessels. Circ. Res. 103, 1092–1099
Received 14 July 2011
doi:10.1042/BST20110652
C
The Authors Journal compilation C
2011 Biochemical Society
... One main approach revolves around the administration and targeting of the pro-angiogenic molecule, vascular endothelial growth factor (VEGF) 5 . While VEGF is a potent inducer of angiogenesis, the success of VEGF-based gene therapies is limited as adequate angiogenic response requires tight control of VEGF level and its duration of effect 6,7 . Low or transient expression of VEGF has minimal angiogenic effects, whereas high or prolonged expression induces dysfunctional vessels with structural abnormalities and may result in the development of angioma. ...
Article
Full-text available
Therapeutic angiogenesis represents a promising avenue to revascularize the ischemic heart. Its limited success is partly due to our poor understanding of the cardiac stroma, specifically mural cells, and their response to ischemic injury. Here, we combine single-cell and positional transcriptomics to assess the behavior of mural cells within the healing heart. In response to myocardial infarction, mural cells adopt an altered state closely associated with the infarct and retain a distinct lineage from fibroblasts. This response is concurrent with vascular rarefaction and reduced vascular coverage by mural cells. Positional transcriptomics reveals that the infarcted heart is governed by regional-dependent and temporally regulated programs. While the remote zone acts as an important source of pro-angiogenic signals, the infarct zone is accentuated by chronic activation of anti-angiogenic, pro-fibrotic, and inflammatory cues. Together, our work unveils the spatiotemporal programs underlying cardiac repair and establishes an association between vascular deterioration and mural cell dysfunction.
... ANG2 -the antagonist of ANG1 -destabilizes established blood vessels and promotes vessel remodeling and angiogenesis (Carmeliet and Jain, 2011;Gerald et al., 2013). There is also crosstalk between the VEGF and ANG2 signaling pathways: VEGF upregulates ANG2 expression in endothelial cells (Reginato et al., 2011) while ANG2 sensitizes endothelial cells to VEGF signaling (Fiedler and Augustin, 2006). Accordingly, combined inhibition of VEGF and ANG2 has demonstrated improved antitumor efficacy versus VEGF or ANG2 inhibition alone in preclinical models (Brown et al., 2010;Hashizume et al., 2010). ...
Article
VEGF and angiopoietin-2 (ANG2) have complementary roles in angiogenesis and promote an immunosuppressive tumor microenvironment. It is anticipated that combination of VEGF and ANG2 blockade could provide superior activity to blockade of either pathway alone, and that addition of VEGF/ANG2 inhibition to an anti-programmed cell death protein-1 (PD-1) antibody could change the tumor microenvironment to support T-cell-mediated tumor cytotoxicity. Here, we describe the pharmacologic and antitumor activity of BI 836880, a humanized bispecific nanobodyTM comprising two single variable domains blocking VEGF and ANG2, and an additional module for half-life extension in vivo BI 836880 demonstrated high affinity and selectivity for human VEGF-A and ANG2, resulting in inhibition of downstream signaling of VEGF/ANG2 and a decrease in endothelial cell proliferation and survival. In vivo, BI 836880 exhibited significant antitumor activity in all patient-derived xenograft models tested, showing significantly greater tumor growth inhibition (TGI) than bevacizumab (VEGF inhibition) and AMG386 (ANG1/2 inhibition) in a range of models. In a Lewis lung carcinoma syngeneic tumor model, combination of PD-1 inhibition with VEGF inhibition showed superior efficacy versus blockade of either pathway alone. TGI was further increased with addition of ANG2 inhibition to VEGF/PD-1 blockade. VEGF/ANG2 inhibition had a strong anti-angiogenic effect. Our data suggest that blockade of VEGF and ANG2 with BI 836880 may offer improved antitumor activity versus blockade of either pathway alone and that combining VEGF/ANG2 inhibition with PD-1 blockade can further enhance antitumor effects. Significance Statement VEGF and ANG2 play key roles in angiogenesis and have an immunosuppressive effect in the tumor microenvironment. Here, we show that BI 836880, a bispecific nanobodyTM targeting VEGF and ANG2, demonstrates substantial antitumor activity in preclinical models. Combining VEGF/ANG2 inhibition with blockade of the PD-1 pathway can further improve antitumor activity.
... 8,9 There is also crosstalk between the two pathways as Ang-2 enhances VEGF signaling, and VEGF upregulates Ang-2 expression. 9,10 VEGF and Ang-2 also have distinct immunosuppressive effects in the tumor microenvironment. 11 Thus, inhibiting VEGF and Ang-2 may also enhance the tumor microenvironment to support T cell function, providing a rationale for combining with agents that target the programmed death protein-1 (PD-1) pathway. ...
Article
Full-text available
Background BI 836880 is a humanized bispecific nanobody® that inhibits vascular endothelial growth factor and angiopoietin-2. Here, we report results from two phase I, nonrandomized, dose-escalation studies (NCT02674152 and NCT02689505; funded by Boehringer Ingelheim) evaluating BI 836880 in patients with confirmed locally advanced or metastatic solid tumors, refractory to standard therapy, or for which standard therapy was ineffective. Patients and Methods Patients aged ≥18 years, with an Eastern Cooperative Oncology Group performance status of 0-2 and adequate organ function received escalating intravenous doses of BI 836880 once every 3 weeks (Q3W; Study 1336.1) or once weekly (QW; Study 1336.6). Primary objectives were maximum tolerated dose (MTD) and recommended phase II dose of BI 836880, based on dose-limiting toxicities (DLTs) during the first cycle. Results Patients received one of five dosages of 40-1000 mg Q3W (29 patients) or 40-240 mg QW (24 patients). One DLT occurred with Q3W treatment [Grade (G) 3 pulmonary embolism (1000 mg)]. Five DLTs occurred in four patients treated QW [G2 proteinuria (120 mg); G3 hypertension (180 mg); G3 proteinuria and G3 hypertension (240 mg); and G4 respiratory distress (240 mg)]. All patients experienced adverse events, most commonly hypertension with Q3W treatment (89.7%; G3 41.4%), and asthenia with QW treatment (62.5%). Two patients treated Q3W (both 1000 mg) and three patients treated QW (120 mg, 2 patients; 180 mg, 1 patient) experienced partial response. Conclusions The MTD of BI 836880 was 720 mg Q3W and 180 mg QW. BI 836880 was generally manageable and demonstrated preliminary efficacy. Clinical trial registration ClinicalTrials.gov NCT02674152; https://clinicaltrials.gov/ct2/show/NCT02674152 and NCT02689505; https://clinicaltrials.gov/ct2/show/NCT02689505
... Vascular maturation and stabilization are the processes by which endothelial cells return to quiescence and independence from continued VEGF signaling and require the physical association of endothelium with a population of mesenchymal cells called pericytes. These are recruited by platelet-derived growth factor-BB (PDGF-BB), produced by activated endothelium, establish tight cell-to-cell contact with endothelial cells by residing under their basal lamina, and regulate their function through a complex cross-talk involving both paracrine and cell contact-dependent signaling pathways (Reginato et al. 2011). An active role in vascular maturation is also played by a recently identified population of myeloid cells (Giacca and Zacchigna 2012), which are recruited by Semaphorin-3a signaling through its receptor Neuropilin-1 and are therefore called neuropilin-expressing monocytes (NEM), and produce a host of pro-maturation paracrine factors, including PDGF-BB, Angiopoietin-1, and transforming growth factor-β1 (TGF-β1) (Groppa et al. 2015). ...
Article
Full-text available
Therapeutic angiogenesis aims at promoting the growth of blood vessels to restore perfusion in ischemic tissues or aid tissue regeneration. Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis in development, repair, and disease. However, exploiting VEGF for therapeutic purposes has been challenging and needs to take into account some key aspects of VEGF biology. In particular, the spatial localization of angiogenic signals within the extracellular matrix is crucial for physiological assembly and function of new blood vessels. Fibrin is the provisional matrix that is universally deposited immediately after injury and supports the initial steps of tissue regeneration. It provides therefore several ideal features as a substrate to promote therapeutic vascularization, especially through its ability to present growth factors in their physiological matrix-bound state and to modulate their availability for signaling. Here, we provide an overview of fibrin uses as a tissue-engineering scaffold material and as a tunable platform to finely control dose and duration of delivery of recombinant factors in therapeutic angiogenesis. However, in some cases, fibrin has also been associated with undesirable outcomes, namely the promotion of fibrosis and scar formation that actually prevent physiological tissue regeneration. Understanding the mechanisms that tip the balance between the pro-and anti-regenerative functions of fibrin will be the key to fully exploit its therapeutic potential.
... 115,121 Finally, for the development of a mature, stable microvessel with a confluent endothelial layer that is viable long-term, coculture with mural cells is also a requirement, which could be implemented by the use of organoids, which already contain microvascular structures composed of both ECs and mural cells, into the hydrogel. 33,46,109,122 4.6. Scaffold Performance. ...
Article
Full-text available
Chronic kidney disease affects one in six people worldwide. Due to the scarcity of donor kidneys and the complications associated with hemodialysis (HD), a cell-based bioartificial kidney (BAK) device is desired. One of the shortcomings of HD is the lack of active transport of solutes that would normally be performed by membrane transporters in kidney epithelial cells. Specifically, proximal tubule (PT) epithelial cells play a major role in the active transport of metabolic waste products. Therefore, a BAK containing an artificial PT to actively transport solutes between the blood and the filtrate could provide major therapeutic advances. Creating such an artificial PT requires a biocompatible tubular structure which supports the adhesion and function of PT-specific epithelial cells. Ideally, this scaffold should structurally replicate the natural PT basement membrane which consists mainly of collagen fibers. Fiber-based technologies such as electrospinning are therefore especially promising for PT scaffold manufacturing. This review discusses the use of electrospinning technologies to generate an artificial PT scaffold for ex vivo/in vivo cellularization. We offer a comparison of currently available electrospinning technologies and outline the desired scaffold properties required to serve as a PT scaffold. Discussed also are the potential technologies that may converge in the future, enabling the effective and biomimetic incorporation of synthetic PTs in to BAK devices and beyond.
Article
Full-text available
Sterile inflammation after injury is important for tissue restoration. In injured human and mouse tissues, macrophages were recently found to accumulate perivascularly. This study investigates if macrophages adopt a mural cell phenotype important for restoration after ischemic injury. Single-cell RNA sequencing of fate-mapped macrophages from ischemic mouse muscles demonstrates a macrophage-toward-mural cell switch of a subpopulation of macrophages with downregulated myeloid cell genes and upregulated mural cell genes, including PDGFRβ . This observation was further strengthened when including unspliced transcripts in the analysis. The macrophage switch was proven functionally relevant, as induction of macrophage-specific PDGFRβ deficiency prevented their perivascular macrophage phenotype, impaired vessel maturation and increased vessel leakiness, which ultimately reduced limb function. In conclusion, macrophages in adult ischemic tissue were demonstrated to undergo a cellular program to morphologically, transcriptomically and functionally resemble mural cells while weakening their macrophage identity. The macrophage-to-mural cell-like phenotypic switch is crucial for restoring tissue function and warrants further exploration as a potential target for immunotherapies to enhance healing.
Preprint
Full-text available
Sterile inflammation following injury is important for tissue restoration. In injured human and mouse tissues, macrophages were recently found to accumulate perivascularly. This study investigates if macrophages adopt a mural cell identity important for restoration following ischemic injury. Single-cell RNA-sequencing of fate-mapped macrophages from ischemic mouse muscles demonstrates an identity switch of a subpopulation of macrophages with downregulated myeloid cell genes and upregulated mural cell genes. This macrophage-to-mural cell switch was further strengthened when including unspliced transcripts in the analysis. Induction of macrophage-specific PDGFRβ-deficiency prevented the perivascular macrophage phenotype, impaired vessel maturation and increased vessel leakiness, which ultimately reduced limb function. In conclusion, macrophages in adult ischemic tissue were demonstrated to undergo a transdifferentiation program to morphologically, transcriptomically and functionally resemble mural cells while losing their macrophage identity. The macrophage-to-mural cell switch is crucial for restored tissue function, and warrants exploration for future immunotherapies to enhance healing following injury.
Article
Full-text available
Chronic ethanol exposure affects the glutamatergic system in several brain reward regions such as the nucleus accumbens (NAc). Our laboratory has shown that chronic exposure to ethanol reduced the expression of glutamate transporter 1 (GLT-1) and cystine/glutamate exchanger transporter (xCT) and, as a result, increased extracellular glutamate concentrations in the NAc of alcohol preferring (P) rats. Moreover, previous study from our laboratory reported that chronic ethanol intake altered the expression of certain metabotropic glutamate receptors in the brain. In addition to central effects, chronic ethanol consumption induced liver injury, which is associated with steatohepatitis. In the present study, we investigated the effects of chronic ethanol consumption in the brain and liver. Male P rats had access to free choice of ethanol and water bottles for five weeks. Chronic ethanol consumption reduced GLT-1 and xCT expression in the NAc shell but not in the NAc core. Furthermore, chronic ethanol consumption increased fat droplet content as well as peroxisome proliferator-activated receptor alpha (PPAR-α) and GLT-1 expression in the liver. Importantly, treatment with the novel beta-lactam compound, MC-100093, reduced ethanol drinking behavior and normalized the levels of GLT-1 and xCT expression in the NAc shell as well as normalized GLT-1 and PPAR-α expression in the liver. In addition, MC100093 attenuated ethanol-induced increases in fat droplet content in the liver. These findings suggest that MC-100093 might be a potential lead compound to attenuate ethanol-induced dysfunction in glutamatergic system and liver injury. Significance Statement This study identified a novel beta-lactam, MC100093, that has upregulatory effects in GLT-1. MC-100093, reduced ethanol drinking behavior and normalized levels of GLT-1 and xCT expression in the NAc shell as well as normalizing GLT-1 and PPAR-α expression in the liver. In addition, MC100093 attenuated ethanol-induced increases in fat droplet content in the liver.
Article
ERG (ETS-related gene) is a member of the ETS (Erythroblast-transformation specific) family of transcription factors abundantly present in vascular endothelial cells. Recent studies demonstrate that ERG has important roles in blood vessel stability and angiogenesis. However, it is unclear how ERG is potentially involved in microvascular barrier functions and permeability. A wide variety of diseases and clinical conditions including trauma-hemorrhagic shock and burn injury are associated with microvascular dysfunctions, which causes excessive microvascular permeability, tissue edema and eventually, multiple organ dysfunction and death. The main purpose of this study was to determine the specific role of ERG in regulating microvascular permeability in human lung microvascular endothelial cells (HLMEC) and to evaluate if exogenous ERG will protect the barrier. The HLMECs were grown on Transwell inserts as monolayers and were transfected with ERG CRISPR/cas9 knockdown plasmid, ERG CRISPR activation plasmid, recombinant ERG protein or their respective controls. Recombinant vascular endothelial growth factor (VEGF) was used as an inducer of permeability for evaluating the effect of ERG activation on permeability. Changes in barrier integrity and permeability were studied using monolayer permeability assay and immunofluorescence of adherens junction proteins (VE-cadherin and β-catenin) respectively. CRISPR/cas9-based ERG knockdown as well as VEGF treatment induced monolayer hyperpermeability, VE-cadherin, and β-catenin junctional relocation and cytoskeletal F-actin stress fiber formation. CRISPR based ERG activation and recombinant ERG transfection attenuated VEGF-induced monolayer hyperpermeability. ERG activation preserved the adherens junctions and cytoskeleton. These results demonstrate that ERG is a potent regulator of barrier integrity and permeability in human lung microvascular endothelial cells and endogenously or exogenously enhancing ERG provides protection against barrier dysfunction and hyperpermeability.
Article
Full-text available
The use of various transgenic mouse models and analysis of human tumour biopsies has shown that bone marrow-derived myeloid cells, such as macrophages, neutrophils, eosinophils, mast cells and dendritic cells, have an important role in regulating the formation and maintenance of blood vessels in tumours. In this Review the evidence for each of these cell types driving tumour angiogenesis is outlined, along with the mechanisms regulating their recruitment and activation by the tumour microenvironment. We also discuss the therapeutic implications of recent findings that specific myeloid cell populations modulate the responses of tumours to agents such as chemotherapy and some anti-angiogenic therapies.
Article
Full-text available
Little is known about how the initial endothelial plexus is remodelled into a mature and functioning vascular network. Studying postnatal remodelling of the retina vasculature, we show that a critical step in vascular maturation, namely pericyte recruitment, proceeds by outmigration of cells positive for (alpha)-smooth muscle actin from arterioles and that coverage of primary and smaller branches lags many days behind formation of the endothelial plexus. The transient existence of a pericyte-free endothelial plexus coincides temporally and spatially with the process of hyperoxia-induced vascular pruning, which is a mechanism for fine tuning of vascular density according to available oxygen. Acquisition of a pericyte coating marks the end of this plasticity window. To substantiate that association with pericytes stabilizes the vasculature, endothelial-pericyte associations were disrupted by intraocular injection of PDGF-BB. Ectopic PDGF-BB caused the detachment of PDGF-beta receptor-positive pericytes from newly coated vessels, presumably through interference with endogenous cues, but had no effect on mature vessels. Disruption of endothelial-pericyte associations resulted in excessive regression of vascular loops and abnormal remodelling. Conversely, intraocular injection of VEGF accelerated pericyte coverage of the preformed endothelial plexus, thereby revealing a novel function of this pleiotropic angiogenic growth factor. These findings also provide a cellular basis for clinical observations that vascular regression in premature neonates subjected to oxygen therapy [i.e. in retinopathy of prematurity] drops precipitously upon maturation of retina vessels and a mechanistic explanation to our previous findings that VEGF can rescue immature vessels from hyperoxia-induced regression.
Article
Full-text available
We set out to investigate the ability of cardiotropic adeno-associated viral vector (AAV2.9 = recombinant adeno-associated virus [rAAV]) to induce prolonged expression of vascular endothelial growth factor (VEGF)-A and platelet-derived growth factor (PDGF)-B in a rabbit hindlimb ischemia model and a pig model of hibernating myocardium. Gene therapy to induce angiogenesis and arteriogenesis has produced mixed results. However, long-acting viruses, such as rAAV, as well as combined induction of angiogenesis and vessel maturation might extend the therapeutic potential. In rabbits, 0.5 x 10(11) particles rAAV.VEGF-A with or without 1 x 10(12) particles rAAV.PDGF-B were retroinfused at day 7 after femoral artery excision. At days 7 and 35, collateral counts and perfusion were determined, each value given as the day 35/day 7 ratio. Capillary-to-muscle fiber ratio was determined at day 35. In pigs, implantation of a reduction stent graft into the circumflex artery led to complete occlusion at day 28. At this time point, retroinfusion of rAAV.VEGF-A (1 x 10(13) particles), rAAV.VEGF-A/PDGF-B (2 x 10(12) and 4 x 10(12) particles, respectively) or mock transfection was performed. Ejection fraction and left ventricular end-diastolic pressure were assessed at days 28 and 56. In rabbits, rAAV.VEGF-A strongly induced angiogenesis (capillary-to-muscle fiber ratio; 1.67 +/- 0.09 vs. 1.32 +/- 0.11 in rAAV.LacZ-treated limbs, p < 0.05), but not collateral growth (125 +/- 7% vs. 106 +/- 7%, p = NS) or perfusion (136 +/- 12% vs. 107 +/- 9%, p = NS). With VEGF-A/PDGF-B cotransfection, collateral growth increased to 146 +/- 9%, perfusion to 163 +/- 8% of the respective day 7 value (p < 0.05). In the pig model, retroinfusion of rAAV.VEGF-A/PDGF-B increased regional myocardial blood flow reserve from 101 +/- 4% (rAAV.Mock) to 129 +/- 8% (p < 0.05), based on collateral growth (3.2 +/- 0.3 in rAAV.Mock vs. 9.0 +/- 0.4 in rAAV.VEGF-A/PDGF-B, p < 0.05), whereas rAAV.VEGF-A did not alter flow reserve (112 +/- 7%) or collateral count (5.2 +/- 0.7). rAAV.VEGF-A/PDGF-B improved ejection fraction (55 +/- 5% vs. 34 +/- 3% in rAAV.Mock, p < 0.05) unlike rAAV.VEGF-A (37 +/- 2%). Retroinfusion of rAAV.VEGF-A alone induces angiogenesis, but fails to enhance collateralization and perfusion, unless PDGF-B is cotransfected. In addition to neovascularization, rAAV.VEGF-A/PDGF-B improves regional and global myocardial function in hibernating myocardium.
Article
Little is known about how the initial endothelial plexus is remodelled into a mature and functioning vascular network. Studying postnatal remodelling of the retina vasculature, we show that a critical step in vascular maturation, namely pericyte recruitment, proceeds by outmigration of cells positive for α-smooth muscle actin from arterioles and that coverage of primary and smaller branches lags many days behind formation of the endothelial plexus. The transient existence of a pericyte-free endothelial plexus coincides temporally and spatially with the process of hyperoxia-induced vascular pruning, which is a mechanism for fine tuning of vascular density according to available oxygen. Acquisition of a pericyte coating marks the end of this plasticity window. To substantiate that association with pericytes stabilizes the vasculature, endothelial-pericyte associations were disrupted by intraocular injection of PDGF-BB. Ectopic PDGF-BB caused the detachment of PDGF-β receptor-positive pericytes from newly coated vessels, presumably through interference with endogenous cues, but had no effect on mature vessels. Disruption of endothelial-pericyte associations resulted in excessive regression of vascular loops and abnormal remodelling. Conversely, intraocular injection of VEGF accelerated pericyte coverage of the preformed endothelial plexus, thereby revealing a novel function of this pleiotropic angiogenic growth factor. These findings also provide a cellular basis for clinical observations that vascular regression in premature neonates subjected to oxygen therapy [i.e. in retinopathy of prematurity] drops precipitously upon maturation of retina vessels and a mechanistic explanation to our previous findings that VEGF can rescue immature vessels from hyperoxia-induced regression.
Article
The aim of this research was to test the effects of vascular endothelial growth factor (VEGF)/angiopoietin-1 (Ang-1) on adult hypoperfused tissues. Angiopoietin-1 and VEGF act separately and synergistically in vascular development during embryogenesis. However, little is known regarding their relative roles in collateral development after chronic arterial obstruction and tissue ischemia in the adult. Central and caudal ear arteries of 32 rabbits were ligated to induce ischemia. At two months, when flow was about 65% of pre-ligation values, we injected intradermally 10 9 plaque-forming unit adenovirus with the following transgenes: Ang-1, VEGF, or a combination of both. Ear perfusion was followed up for four weeks, and vessel leakage was assessed by Evens Blue test. Before injection, flow was 65% of baseline, and endogenous VEGF levels in ischemic tissue were increased. Adenovirus-encoding VEGF gene (Ad.VEGF) at one week caused a visible inflammatory response associated with a 24% flow increase (p = 0.018). Adenovirus-encoding Ang-1 gene (Ad.Ang-1) increased flow 22% (p = 0.004) with no visible inflammation; Ad.VEGF caused three times as much vessel leakage as Ad.Ang-1 (142.5 ± 38 vs. 49.5 ± 9.8 ng Evens Blue/mg tissue; p < 0.001). However, at four weeks, compared with baseline, VEGF decreased flow 18% (p = 0.004), whereas Ang-1 increased tissue perfusion 26% (p < 0.001). This effect was abolished when Ad.Ang-1 was injected with soluble VEGF receptor [Ad.Flt(1-3)-Fc], which blocks VEGF-dependent signaling. Exogenous Ang-1 did not increase perfusion in a normally perfused ear, in which endogenous VEGF is not expressed. Exogenous Ang-1 enhances perfusion in hypoperfused tissues only in the presence of increased levels of endogenous VEGF. Overexpression of VEGF, however, after causing an inflammatory response, does not improve collateral blood flow.
Article
Constitutive expression of VEGF after implantation of genetically engineered myoblasts into non-ischemic muscle led to an increase in vascular structures. Previously, effects of VEGF delivery to adult muscle have only been reported in ischemic tissues. The resulting vascular structures were reminiscent of those formed during embryonic vasculogenesis, rather than angiogenesis, sprouting from preexisting vessels. Initially, VEGF caused an accumulation of endothelial cells and macrophages, followed by networks of vascular channels and hemangiomas with locally high serum VEGF levels. No effects were evident in adjacent tissue or contralateral legs, where low serum VEGF was detected. These data suggest that the induction by VEGF of angiogenesis or vasculogenesis may be dose-dependent. Furthermore, VEGF expression must be carefully modulated, as overexpression is deleterious.
Article
Angiopoietin-1 (Ang1) and vascular endothelial growth factor (VEGF) are endothelial cell–specific growth factors. Direct comparison of transgenic mice overexpressing these factors in the skin revealed that the VEGF-induced blood vessels were leaky, whereas those induced by Ang1 were nonleaky. Moreover, vessels in Ang1-overexpressing mice were resistant to leaks caused by inflammatory agents. Coexpression of Ang1 and VEGF had an additive effect on angiogenesis but resulted in leakage-resistant vessels typical of Ang1. Ang1 therefore may be useful for reducing microvascular leakage in diseases in which the leakage results from chronic inflammation or elevated VEGF and, in combination with VEGF, for promoting growth of nonleaky vessels.
Article
Blood vessels deliver oxygen and nutrients to every part of the body, but also nourish diseases such as cancer. Over the past decade, our understanding of the molecular mechanisms of angiogenesis (blood vessel growth) has increased at an explosive rate and has led to the approval of anti-angiogenic drugs for cancer and eye diseases. So far, hundreds of thousands of patients have benefited from blockers of the angiogenic protein vascular endothelial growth factor, but limited efficacy and resistance remain outstanding problems. Recent preclinical and clinical studies have shown new molecular targets and principles, which may provide avenues for improving the therapeutic benefit from anti-angiogenic strategies.
Article
Tightly regulated crosstalk between endothelial cells and pericytes is required for formation and maintenance of functional blood vessels. When the NG2 proteoglycan is absent from pericyte surfaces, vascularization of syngeneic tumors growing in the C57Bl/6 mouse brain is aberrant in several respects, resulting in retardation of tumor progression. In the NG2 null mouse brain, pericyte investment of the tumor vascular endothelium is reduced, causing deficiencies in both pericyte and endothelial cell maturation, as well as reduced basal lamina assembly. While part of this deficit may be due to the previously-identified role of NG2 in beta1 integrin-dependent periyte/endothelial cell crosstalk, the ablation of NG2 also appears responsible for loss of collagen VI anchorage, in turn leading to reduced collagen IV deposition. Poor functionality of tumor vessels in NG2 null brain is reflected by reduced vessel patency and increased vessel leakiness, resulting in large increases in tumor hypoxia. These findings demonstrate the importance of NG2-dependent pericyte/endothelial cell interaction in the development and maturation of tumor blood vessels, identifying NG2 as a potential target for anti-angiogenic cancer therapy.