You are on page 1of 14

DIABETES/METABOLISM RESEARCH AND REVIEWS REVIEW ARTICLE

Diabetes Metab Res Rev 2003; 19: 442–455.


Published online in Wiley InterScience (www.interscience.wiley.com). DOI: 10.1002/dmrr.415

Vascular endothelial growth factor and diabetic


retinopathy: pathophysiological mechanisms
and treatment perspectives

Ruth B. Caldwell1,2,4 * Summary


Manuela Bartoli1,5
M. Ali Behzadian1,3 Retinal neovascularization and macular edema are central features of
Azza E. B. El-Remessy1,3 diabetic retinopathy, the major cause of blindness in the developed world.
Current treatments are limited in their efficacy and are associated with
Mohamed Al-Shabrawey1
significant adverse effects. Characterization of the molecular and cellular
Daniel H. Platt1 processes involved in vascular growth and permeability has led to the
R. William Caldwell3 recognition that the angiogenic growth factor and vascular permeability
1
factor vascular endothelial growth factor (VEGF) plays a pivotal role in the
Vascular Biology Center, The
retinal microvascular complications of diabetes. Therefore, VEGF represents
Medical College of Georgia Augusta,
an exciting target for therapeutic intervention in diabetic retinopathy. This
GA 30912
review highlights the current understanding of the mechanisms that regulate
2
Department of Cellular Biology and VEGF gene expression and mediate its biological effects and how these
Anatomy, The Medical College of
processes may become altered during diabetes. The cellular and molecular
Georgia Augusta, GA 30912
alterations that characterize experimental models of diabetes are considered
3
Department of Pharmacology and in relation to the influence of high glucose-mediated oxidative stress on VEGF
Toxicology, The Medical College of expression and on the mechanisms of VEGF’s actions under hyperglycemic
Georgia Augusta, GA 30912
induction. Finally, potential therapeutic strategies for preventing VEGF
4
Department of Ophthalmology, The overexpression or blocking its pathological effects in the diabetic retina
Medical College of Georgia Augusta, are considered. Copyright  2003 John Wiley & Sons, Ltd.
GA 30912
5
Department of Pathology, The Keywords diabetic retinopathy; retina; vascular endothelial growth factor;
Medical College of Georgia Augusta, angiogenesis; vascular permeability; reactive oxygen species
GA 30912

*Correspondence to:
Ruth B. Caldwell, Vascular Biology Introduction
Center, Department of Cellular
Biology and Anatomy, Department Clinical features of diabetic retinopathy
of Ophthalmology, The Medical
College of Georgia, Augusta, GA Diabetic retinopathy is the most common complication of diabetes and a
30912. E-mail: rcaldwel@mcg.edu leading cause of blindness in the western world, affecting approximately
three-fourths of diabetic patients within 15 years after onset of the disease
[1–3]. Although the best way to avoid visual loss is to initiate treatment
before symptoms develop, many diabetic patients are referred for treatment
only after visual complications have already occurred. Hyperglycemia is
known to be the primary pathogenic factor in the development of diabetes
complications [4,5], but the mechanisms by which elevated blood glucose
causes tissue injury and disease progression in the retina are not yet clear.
The clinical progression of diabetic retinopathy follows a pattern
characteristic of ischemic retinopathy (for review, see [6,7]). The disease
Received: 23 April 2003
begins with biochemical and cellular alterations that are not clinically
Revised: 12 August 2003
evident. Vascular alterations during this period include increased adhesion
Accepted: 18 August 2003
of leukocytes to the vessel wall, alterations in blood flow, death of retinal

Copyright  2003 John Wiley & Sons, Ltd.


VEGF and Diabetic Retinopathy 443

pericytes (perivascular contractile cells), basement mem- analog had beneficial effects at near toxic doses, whereas,
brane thickening and subtle increases in vascular perme- a receptor blocker was without effect.
ability. Blockage of the retinal capillaries causes localized Epidemiologic observations have suggested that the risk
hypoxia, which is thought to lead to increased tissue pro- and/or progression of diabetic retinopathy is increased by
duction of angiogenic factors. This growth factor release hypertension. Studies in which strict control of blood
is likely to be involved in the permeability increase pressure was obtained using an angiotensin converting
and may contribute to the pathological vascular growth enzyme (ACE) inhibitor or a beta adrenergic blocker
that is seen later. With time, endothelial cells lining found reductions in the progression of retinopathy and
the microvessels are lost, leading to capillary closure risk of deterioration in visual acuity [9]. Because the
and formation of acellular, nonperfused vessels. As the previous studies were not specifically designed to study
disease progresses, obvious alterations in the vascular retinopathy, a new study is underway to see whether
structure can be seen upon ophthalmoscope examination. blockade of the renin-angiotensin system with the AT1-
These include microaneurysms, dot/blot hemorrhages, receptor blocker candesartan can prevent the incidence
cotton-wool spots, venous beading and vascular loops. At and progression of retinopathy.
this stage, the blood vessels become prominently leaky, Another promising therapy in clinical trials is the use
allowing blood and fluid to accumulate in the retinal tis- of oral inhibitors of the beta isoform of protein kinase
sue, forming exudate deposits. The retinal edema causes C (PKCβ). Studies in a variety of experimental models
reduced vision if the macula is affected, but is other- in vivo and in vitro have demonstrated the involvement of
wise asymptomatic. The condition of macular edema PKCβ in the very early ocular complications of diabetes
is also referred to as nonproliferative retinopathy. In (for review, see [10]). These observations have led to the
many patients, the retinopathy progresses to proliferative initiation of clinical trials testing the ability of the specific
retinopathy, which is characterized by the growth of new PKCβ inhibitor LY333531 to slow or halt the development
blood vessels on the surface of the retina. The walls of of proliferative diabetic retinopathy and reduce or reverse
the new blood vessels are weak and may break, allow- the progression of diabetic macular edema (for review,
ing blood to leak out. This can cloud the vitreous and see [8,11]).
compromise vision. In more advanced stages of prolifera- A therapeutic strategy for treating diabetic retinopathy
tive retinopathy, newly formed fibro-vascular tissue grows just beginning to be tested in clinical trials involves
from the retinal surface into the vitreous cavity. This can the use of agents that directly inhibit angiogenesis.
cause retinal detachment and will result in blindness if Until recently, the major emphasis in clinical trials
untreated. Proliferative retinopathy typically develops in of antiangiogenic therapies has been on determining
patients with type 1 diabetes, whereas nonproliferative their ability to inhibit the growth of vascularized
retinopathy with macular edema is more common in tumors [12,13]. However, several agents found to be
patients with type 2 diabetes. effective in controlling tumor growth have now been
evaluated for their efficacy in the treatment of ocular
neovascularization associated with age-related macular
Current therapy for diabetic degeneration [14,15]. The results may have implications
retinopathy for other kinds of pathological neovascularization,
including diabetic retinopathy. Interferon alpha-2a, which
Many therapies for diabetic retinopathy have been eval- had been shown to be antiangiogenic in animal and
uated clinically (for review, see [8]). Laser photocoag- in vitro models, was found to be ineffective in halting
ulation has long been the recommended treatment for the progression of neovascular macular degeneration
patients with advanced proliferative diabetic retinopathy in a double-blind, placebo-controlled clinical trial [16].
or clinically significant diabetic macular edema. While Thalidomide is another promising antiangiogenic drug
this treatment is usually effective, the procedure destroys that has been tested for patients with age-related macular
neural tissue and can decrease peripheral vision, impair degeneration in phase 3 clinical trials, but the results
night vision and change color perception. Moreover, in are not yet known. To date, the most promising results
some patients the retinopathy continues to progress in for treating age-related macular degeneration have been
spite of appropriate treatment. Therefore, there is a great obtained with antiangiogenic reagents targeting vascular
need for the development of new noninvasive therapies endothelial growth factor (VEGF) [8]. The discussion that
to prevent and treat diabetic retinal vascular disease. follows focuses on VEGF, its mechanisms of actions under
The Early Treatment Diabetic Retinopathy Study hyperglycemic induction, its role in diabetic retinopathy
evaluated aspirin treatment and found no improvement and potential strategies for blocking its adverse affects on
of visual acuity in patients with macular edema and the retinal vasculature during diabetes.
no change in the subsequent development of severe
preproliferative diabetic retinopathy. Clinical trials of
aldose reductase inhibitors also showed no significant Hyperglycemia in diabetic retinopathy
effect on the development or progression of diabetic
retinopathy. Small-scale studies showed that blocking the As outlined above, type 1 and type 2 diabetes differ
action of growth hormone using a synthetic somatostatin in manifestation of retinal complications. However, the

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
444 R. B. Caldwell et al.

microvascular alterations in both conditions have the Peroxynitrite is a highly reactive oxidant formed by the
same pathophysiological basis in that the occurrence combination of nitric oxide with superoxide. Research
and progression of retinopathy is closely correlated with demonstrating the presence of the peroxynitrite marker
the degree of hyperglycemia. Clinical trials have shown nitrotyrosine in placental vessels and plasma of diabetic
that intensive blood glucose control can delay the onset patients and retinas of diabetic rats suggests a role for
and reduce the severity of the retinal complications in peroxynitrite in the development of diabetic complications
patients with either type 1 or type 2 diabetes [4,5]. [30–32]. A working model to explain the potential
In other words, the initial stages of retinopathy that interactions between oxidative and nitrative stress in
are collectively related to endothelial dysfunction (i.e. the development of diabetic vascular disease is shown
leukocyte adhesion, blood flow alterations, capillary in Figure 1. This model is based on studies of cultured
closure and formation of leaky vessels) are most probably endothelial cells showing that high glucose-induced
due to the effects of elevated blood glucose concentration superoxide formation leads to an increase in intracellular
on microvascular tissues. calcium, which not only causes activation of endothelial
Numerous studies in various models indicate that nitric oxide synthase (NOS3) to form nitric oxide but also
hyperglycemia in vivo or high glucose exposure in vitro causes NOS3 ‘uncoupling’, leading to further increases
induces the formation of angiogenic factors (for review, in superoxide formation [33–36]. Superoxide combines
see [17,18]). While multiple growth factors and cytokines rapidly with nitric oxide to form peroxynitrite, thereby
have been implicated in retinal neovascular diseases, causing deactivation of nitric oxide. Data showing a
VEGF has been identified as a primary mediator of the reduction in nitric oxide bioavailability and vasodilatory
vascular alterations in diabetic retinopathy (for review, function in endothelial cells exposed to high glucose
see [15,19]). Expression of VEGF is increased by hypoxia, provide further support for this concept [34,35].
oxidative stress, high glucose and inflammatory reactions. Peroxynitrite initiates a variety of pathological pro-
Furthermore, retinal endothelial cells have large numbers cesses including inhibition of key metabolic enzymes
of high affinity, high sensitivity VEGF receptors which by nitration of protein tyrosine residues, lipid perox-
have been found to be increased in diabetes [20,21]. idation and reduction of cellular antioxidant defenses
by oxidation of thiol pools and induction of DNA
strand breaks, leading to apoptosis [37–39]. Peroxynitrite
Oxidative stress in diabetic retinopathy can also oxidize specific cellular components, including
the nitric oxide synthase cofactor tetrahydrobiopterin
The molecular mechanisms of hyperglycemia-induced and may also affect the L-arginine transporter [40,41].
endothelial dysfunction are not fully understood. How- Reduction in availability of tetrahydrobiopterin and/or
ever, multiple biochemical pathways associated with L-arginine uncouples nitric oxide synthase, leading to fur-
hyperglycemia/diabetes-induced vascular injury can ther increases in superoxide and peroxynitrite. This cycle
increase the production of reactive oxygen species. These can be interrupted by scavenging superoxide or perox-
include glucose auto-oxidation, the polyol pathway and ynitrite or inhibiting nitric oxide synthase. Studies in
formation of advanced glycation end products. Moreover, animal models of diabetes and cultured endothelial cells
all of these mechanisms seem to reflect a hyperglycemia- exposed to high glucose have demonstrated that increases
induced process initiated by superoxide overproduction in peroxynitrite formation are directly correlated with dia-
by the mitochondrial electron-transport chain (for review, betes and high glucose-induced VEGF expression and with
see [22]). Recent studies showing that diabetes-induced increases in retinal vascular permeability and reduced
retinal vascular dysfunction can be prevented by inhibitors endothelial cell survival [30,36,42,43].
of reactive oxygen species provide further support for the
role of oxidative stress in diabetic retinopathy [23,24].
Oxidative stress has been correlated with the increased
production of VEGF under in vitro conditions and is
Overview of VEGF actions and its
thought to be involved in the upregulation of VEGF potential role in diabetic retinopathy
expression during diabetes [5,25–27]. Studies showing
that diabetes-induced increases in both retinal VEGF VEGF, a multifunctional vasoactive
concentrations and in lipid peroxidation in streptozotocin- factor
diabetic rats can be prevented by antioxidant treatment
[28] provide strong support for this hypothesis. VEGF is a member of a large family of angiogenic
The mechanisms by which oxidative stress contributes growth factors. There are six known members of the
to diabetic vascular dysfunction and VEGF overexpression VEGF family: VEGF-A (commonly referred to as VEGF),
are complex. It has been shown that inhibitors of placental growth factor, VEGF-B, VEGF-C, VEGF-D and
nitric oxide synthase prevent diabetes-induced vascular VEGF-E, for review, see [44]).
dysfunction in rats [24,29,30], indicating that formation VEGF was first discovered as vascular permeability
of reactive nitrogen species plays a role in the factor, but subsequently was recognized as an angiogenic
pathology. Recently, attention has been focused on the factor and as a specific mitogen for vascular endothelial
role of peroxynitrite in diabetic vascular dysfunction. cells (for review, see [45]). VEGF’s mitogenic activity

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
VEGF and Diabetic Retinopathy 445

High Glucose

Electr NAD(P)H
on
c Oxidase RAGE

Tr n
ha
ans
i
Aldose

port
reductase

PKC
Ca 2+ i
O2−

NOS3 Uncoupling
L-Arginine ONOO −
Tetrahydrobiopterin

NO

Figure 1. Mechanisms of high glucose-induced formation of reactive oxygen and nitrogen species, including superoxide (O2 − ),
nitric oxide (NO) and peroxynitrite (ONOO-)

is demonstrated in vascular and lymphatic endothelial prevent the diabetes-induced permeability, indicating a
cells, but is not seen at appreciable levels in other direct role for VEGF in this pathology [58].
cell types. VEGF also functions as a vasodilator, Because of the lack of rodent models for proliferative
a promoter of endothelial cell migration and an diabetic retinopathy, considerable research has been done
antiapoptotic, endothelial cell survival factor [46–48]. in neonatal animals in which retinal neovascularization
Studies done in vitro have demonstrated VEGF mRNA is induced by hyperoxia exposure. Using these models
and protein expression by numerous retinal cell types, it has been shown that neovascularization is closely
including retinal-pigmented epithelial cells, pericytes, associated with increases in VEGF production and that
astrocytes, Muller glia and endothelial cells [49–52]. the pathological vascular growth can be prevented by
VEGF expression in cultured cells is substantially treatment with selective VEGF inhibitors [49,59–63].
increased by hypoxia [49,52,53] and inflammatory
mediators [54–56], all of which are increased by
oxidative stress. In vitro studies also indicate that Regulation of VEGF gene expression
high glucose-mediated oxidative stress increases the
autocrine expression of VEGF within retinal microvascular Altered VEGF expression has profound pathological
endothelial cells [57]. consequences. Remarkably, the disruption of a single
VEGF allele results in embryonic lethality due to
severe vascular abnormalities, thus indicating that
Role of VEGF in diabetic retinopathy adequate VEGF expression levels are critical for vascular
development [64]. Conversely, the need for tight control
A number of clinical studies have shown a strong of VEGF expression is evident in the pathological
correlation between increases in intraocular VEGF consequences of VEGF overexpression during ischemic
concentration and the development of proliferative retinopathy (for review, see [15,19]).
diabetic retinopathy (for review, see [15]). VEGF may Tissue hypoxia upregulates VEGF by increasing both
also have a key role in the pathogenesis of diabetic the expression and stability of VEGF mRNA [65].
macular edema due to its action in increasing vascular Hypoxic transcriptional regulation of VEGF is mediated
permeability. Studies in streptozotocin diabetic rats at least in part by the action of the heterodimeric
have shown that retinal vascular alterations similar to complex, hypoxia-inducible factor-1 [66]. Hypoxia-
background diabetic retinopathy in patients are associated inducible factor-1 is composed of the constitutively
with increases in retinal VEGF levels and increased produced hypoxia-inducible factor-1β subunit and the
expression of VEGFR2 [20,21]. Inhibition of VEGF can inducible component hypoxia-inducible factor-1α [67].

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
446 R. B. Caldwell et al.

Hypoxia-inducible factor-1α complexes with hypoxia- due to the failure to develop a normal vasculature
inducible factor-1β and is translocated to the nucleus [93,94]. VEGFR2 null mice exhibit altered angioblast
where it binds the hypoxia response elements to promote differentiation, indicating VEGFR2’s involvement in
the expression of VEGF, erythropoietin, and other genes endothelial cell lineage determination. By contrast,
whose products are involved in oxygen transport and VEGFR1 null mice develop abnormal vascular-like
glucose metabolism [68]. The role of hypoxia-inducible structures in which the lumen is filled with endothelial
factor-1α in diabetic retinopathy is not yet clear, but cells, indicating a role for VEGFR1 as negative modulator
it has been implicated in VEGF overexpression and of vascular formation [95]. Overexpression of a short
hyperpermeability of the blood-retinal barrier in diabetic soluble form of VEGFR1 results in negative regulatory
animals treated with intensive insulin therapy during early effects. Finally, recent evidence has implicated VEGFR1
diabetes [69]. in cell migration and in the induction of stem cell
Of particular interest in relation to VEGF overexpression recruitment and vasculogenesis in various models of
during diabetic retinopathy are the effects of TGF-β, TNF- ischemic retinopathy [96,97], suggesting a role for
α, IGF-1, interleukins, advanced glycation end products VEGFR1 in postnatal vasculogenesis. Involvement of
and reactive oxygen species, all of which are upregulated VEGFR1 in diabetic retinopathy remains to be determined.
in diabetes [70–76]. The influence of these factors on VEGFR2 plays a prominent role in mediating VEGF’s
transcriptional events leading to VEGF overexpression angiogenic actions. Upon VEGF binding, VEGFR2 under-
is still not completely understood. However, oxidative goes dimerization and transphosphorylation followed by
stress has been found to increase VEGF expression by a complex cascade of signaling events leading to multiple
a mechanism involving signal transducer and activator cell responses, including vasodilation, increased vascular
of transcription factor 3 (STAT3) [77]. STAT proteins permeability and increases in endothelial cell migra-
are latent cytoplasmic transcription factors that regulate tion, proliferation and survival (Figure 2, for review, see
gene expression induced by cytokines, interferons and [98]). In diabetic retinas, VEGFR2 expression is increased
growth factors (for review, see [78–80]). STAT3 binding and the intracellular signaling events associated with its
sites have been identified in the VEGF promoter, and activation play a significant role in the onset and/or pro-
mutations of these regions can block VEGF mRNA gression of retinopathy [20,21,99]. Thus, VEGFR2 and its
transcription [81]. STAT3 has been shown to play a downstream mediators represent important therapeutic
pivotal role in tumor and heart angiogenesis by inducing targets.
VEGF expression. STAT3 is activated by reactive oxygen Recent studies have indicated that VEGF activation
species as well as by inflammatory mediators, and its of VEGFR2 involves formation of superoxide anion
constitutive activation has been shown to correlate with in endothelial cells through a mechanism involving
increased rates of VEGF expression and angiogenesis in activation of NAD(P)H oxidase. This effect has been
in vivo and in vitro [81–85]. It has been shown that VEGF shown to be required for VEGF’s mitogenic properties
selectively induces STAT3 activation and VEGF autocrine [76], indicating that superoxide can serve as an
expression in retinal microvascular endothelial cells, intracellular signaling mediator of angiogenesis. In
whereas macrovascular endothelial cells from the aorta addition, this result implies that VEGF stimulation of
are unaffected [86,87]. Studies now in progress indicate VEGFR2 is itself a potential source of oxidative stress in
that STAT3 also plays a fundamental role in inducing diabetic retinopathy.
VEGF expression in retinal endothelial cells exposed to
oxidative stress involving peroxynitrite formation [88].
Mechanisms of VEGF-induced increases
in vascular permeability
VEGF receptors
Early breakdown of the blood-retinal barrier in diabetes
Two high-affinity VEGF receptors, VEGFR1 (Flt-1 or at the level of the retinal vascular endothelium has been
fms-like tyrosine kinase) and VEGFR2 (Flk-1/KDR or well established by analyses in both diabetic patients and
fetal liver kinase) have been identified in vascular experimental animals (for review, see [100]).
endothelial cells [89–92]. These receptors are similar in The permeability defect has been shown to correlate
structure – both are class III receptor tyrosine kinases with with increases in the expression of VEGF [58,101–103].
seven immunoglobulin-like loops in their extracellular Recent studies in streptozotocin diabetic rats have shown
domain and a kinase insert region in the intracellular that the initial breakdown of the blood-retinal barrier
domain – but they differ in their biological activities. is associated with increases in expression of the both
VEGFR1 displays a higher ligand affinity than VEGFR2 endothelial and neuronal nitric oxide synthase as well
[89,92]. However, studies with cultured endothelial cells as with increases in VEGF expression [30,103,104].
indicate that lack of VEGFR1 has little effect on VEGF Moreover a VEGF-neutralizing receptor construct has
function, whereas lack of VEGFR2 prevents VEGF-induced been shown to prevent the diabetes-induced increases in
cellular responses (for review, see [45]). expression of VEGF and endothelial nitric oxide synthase
Studies with knockout mice revealed that absence of and the barrier breakdown [104], underlining a potential
either VEGFR1 or VEGFR2 results in embryonic lethality role for VEGF in the early vascular dysfunction.

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
VEGF and Diabetic Retinopathy 447

The specific mechanism of the VEGF-induced bar- oxide synthase or scavenging peroxynitrite, indicating a
rier dysfunction has been controversial, but studies of role for nitric oxide synthase and peroxynitrite formation
VEGF’s permeability-inducing actions in vitro have shown in the pathology.
that VEGF induces a transient and reversible hyper- The identification of mediators that regulate VEGF
permeability in cultured endothelial cells that persists effects on blood/tissue barrier breakdown may have sig-
for only 30 min, which is associated with the for- nificant clinical implications. In vitro analyses of the sig-
mation of fenestrae, a transcellular permeability route naling pathways by which VEGF stimulates the autocrine
[105]. In vitro studies with cultured retinal microvascular production of nitric oxide and prostacyclin by vascular
endothelial cells have shown that this immediate perme- endothelial cells indicate that VEGF activation of VEGFR2
ability increase occurs by a mechanism of transcytosis involves the formation of protein complex between
in caveolin-coated vesicles requiring activation of nitric VEGFR2 and c-Src and that c-Src activation is required
oxide synthase and nitric oxide formation [106]. for VEGF induction of nitric oxide and prostacyclin for-
It was subsequently found that the rapid and transient mation [110] (Figure 2). Further studies have shown that
permeability increase is followed by a sustained increase VEGF induces uPAR gene expression in cultured retinal
in paracellular permeability starting 4 to 6 h after VEGF endothelial cells by inducing transcriptional activation
treatment [107]. Since VEGF induces urokinase receptor of β-catenin and that the delayed paracellular phase of
(uPAR) expression [108], which may initiate plasmin the VEGF-induced permeability increase correlates with
formation, it appears likely that the VEGF-induced uPA/uPAR activation [107]. These findings may shed new
permeability results from disruption of endothelial light on the mechanism of VEGF-induced barrier dysfunc-
cell-to-cell attachments by uPAR-mediated initiation tion. Further definition of the VEGF-induced signaling
of proteolytic activities. This would generate leaky pathways leading to these events should aid in the devel-
vessels and allow endothelial cells to penetrate their opment of more specific and efficient therapies for the
underlying basement membrane, proliferate and migrate, treatment of diabetic macular edema and prevention of
proliferative retinopathy.
setting the stage for retinal neovascularization. These
in vitro results are consistent with results of studies in
streptozotocin-induced diabetic rats showing increased VEGF’s angiogenic actions
vascular permeability associated with reductions in
levels of the tight junction protein occludin [100,109]. VEGF’s functions in stimulating endothelial cell migration
Subsequent work has shown that this diabetes-induced and proliferation during angiogenesis have been well
increase in vascular permeability is associated with established (for review, see [45,111]) and the role of
overexpression of VEGF and uPAR and with oxidative these functions in proliferative diabetic retinopathy is
stress as evidenced by increased formation of nitric oxide, also clear. However, there is increasing evidence that
superoxide and peroxynitrite [30]. Moreover, each of neovascularization is not only due to angiogenesis but
these alterations could be prevented by inhibiting nitric may also involve the process of vasculogenesis in which

VEGF

VEGFR2

P P
Src P
P β - catenin
PI3K FAK
PlCγ
STAT1 uPAR
PKB/Akt &
IP3 DAG STAT3 Paxilin

Survival NOS Ca ++ PKC MAPK Permeability


Gene
NO PGI2
expression Cytoskeletal
rearrangement
Cell proliferation/permeability Cell migration
Vasodilation

Angiogenesis

Figure 2. Signaling pathways activated by VEGF

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
448 R. B. Caldwell et al.

circulating bone marrow-derived endothelial progenitor diabetes in causing pathological overgrowth of the retinal
cells are recruited into the neovascularization process microvasculature are due in part to VEGF’s autocrine
[112]. Relatively little is known yet about the effects actions in triggering VEGF overexpression in retinal
of diabetes on vasculogenesis and the recruitment of microvascular endothelial cells. Parallel actions of high
endothelial precursor cells. However, recent studies glucose/oxidative stress in inhibiting the production of
indicate that vasculogenesis is impaired in a mouse model proteins that normally inhibit VEGF expression may
of diabetes [113]. Studies of human endothelial precursor enhance this autocrine process. This hypothesis is
cells isolated from diabetic patients showed impairments supported by studies showing that decreased endostatin
in their rate of proliferation, adhesion to endothelium levels are correlated with increased VEGF levels in
and incorporation into vascular structures, suggesting patients with proliferative diabetic retinopathy [120]
that alteration in endothelial precursor cell function may and that increased intraocular expression of endostatin
account for impaired growth of the peripheral vasculature reduces VEGF-induced retinal vascular permeability and
during diabetes [114]. The effects of diabetes on inhibits retinal neovascularization in transgenic mice with
endothelial precursor cell recruitment and vasculogenesis overexpression of VEGF in the retina [121].
in retina have not yet been studied. However, recent
studies in a mouse model of ischemic retinopathy have
shown that bone marrow–derived hematopoietic stem VEGF’s prosurvival actions
cells participate in retinal neovascularization [96]. Studies
have also shown that these cells can be used to rescue VEGF’s function as a survival factor for endothelial cells
and stabilize the degenerating vasculature in a mouse has been clearly documented (for review, see [15]).
model of retinal degeneration or to target and inhibit Paradoxically, even though levels of both VEGF and
new vessel formation using a cell-based gene therapy in a VEGFR2 are substantially increased in diabetic retinas,
mouse model of retinopathy of prematurity [115]. Thus, VEGF’s prosurvival function is compromised as shown by
it is possible that alteration in endothelial precursor cell the fact that retinal capillary dropout occurs at the same
function could be involved in the capillary dropout stage time that expression of VEGF and VEGFR2 is increased
of diabetic retinopathy. Further study is needed to directly [15,122]. Thus, alteration in VEGF protein expression
test this hypothesis. Unfortunately, however, research in does not fully explain disease progression in diabetic
this area will be seriously limited until good models for retinopathy. The VEGF prosurvival signaling pathway in
diabetic retinopathy become available. endothelial cells may also be altered during diabetes.
VEGF activation of VEGFR2 is known to result
in the transduction of antiapoptotic signals via the
VEGF autocrine induction of VEGF phosphatidylinositol 3 -kinase/Akt signaling pathway
expression in activated endothelial [123,124]. However, VEGF also stimulates activation of
cells the stress-activated serine/threonine protein kinase, p38
mitogen activated protein kinase [125] – an important
VEGF expression has been described mainly as a modulator of the proapoptotic process in various cell
paracrine event. However, the development of new types, including endothelial cells. Blockade of VEGF-
capillary networks during neovascularization may require mediated activation of phosphatidylinositol 3 -kinase or
the existence of autocrine growth stimulation. VEGF’s Akt signaling has been shown to result in increased
autocrine ability to stimulate its own production in apoptosis due to potentiation of p38 mitogen activated
the microvascular endothelium has been described in protein kinase activation [126]. Studies in retinal
hypoxia, brain tumors, when the cell-to-cell junctions endothelial cells cultured in high glucose medium
are disrupted, or during in vitro angiogenesis induced or treated with peroxynitrite have shown a similar
by advanced glycation end products [84,116]. The role phenomenon of accelerated apoptosis even in the
of VEGF autocrine expression in diabetic retinopathy presence of exogenous VEGF [42,127]. This pro-apoptotic
is not yet known, but high glucose and peroxynitrite effect is associated with increases in phosphorylation
both appear to induce VEGF autocrine expression in of P38 mitogen activated protein kinase, decreases
cultured retinal endothelial cells [57,88]. Diabetes has in Akt-kinase activity and tyrosine nitration of the
also been found to induce the production of inflammatory P85 and P110 subunits of phoshatidylinositol 3 -kinase,
mediators via the cyclooxygenase pathway in the rat retina suggesting a key role for peroxynitrite in high glucose
[117,118]. Products of arachidonic acid metabolism effects impairing endothelial cell survival [42]. Previous
via cyclooxygenase (prostaglandins, prostacyclin and work in vitro has shown that the p85 regulatory
thromboxane) are known to play a role in ocular subunit of phoshatidylinositol 3 -kinase is a target for
inflammation and are able to induce VEGF expression peroxynitrite-induced protein nitration on tyrosine and
in vitro [54–56]. VEGF itself has been shown to induce that tyrosine nitration of p85 blocks its interaction
prostacyclin formation in cultured endothelial cells and with the p110 catalytic subunit [128], indicating that
ICAM-1 expression under both in vivo and in vitro peroxynitrite can alter cell survival responses mediated by
conditions, resulting in amplification of the inflammatory phoshatidylinositol 3 -kinase. Observations of increased
process [110,119]. It appears likely that the effects of tyrosine nitration in retinal proteins of diabetic rats

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
VEGF and Diabetic Retinopathy 449

suggest that peroxynitrite formation is likely to play an targeting VEGF mRNA. Ribozymes are naturally occurring
important role in mediating the suppressive effects of or engineered RNAs with enzymatic activity for degrading
diabetes on endothelial cell survival [30,36]. Peroxynitrite mRNA. The ribozyme, angiozyme, specifically targets
may be ultimately responsible for the pathophysiology and cleaves VEGF receptor mRNA and has been
previously attributed to nitric oxide alone [129,130]. shown to inhibit angiogenesis in animal tumor models
[140].
In addition to the above approaches, a number of
Therapeutic strategies for preventing small molecules have been synthesized that inhibit
VEGF receptor tyrosine kinase activity. SU5416 inhibits
VEGF’s pathological effects VEGFR2 and VEGFR1 intracellular signaling and has
had promising clinical results in inhibiting tumor growth
Inhibitors of VEGF and VEGFRs [141]. It also has some activity against platelet-derived
growth factor receptor, the VEGF receptor Flt-4 and c-
VEGF and its receptors are good targets for ther- kit [142]. ZD4190 is a potent inhibitor of VEGFR1 and
apeutic intervention in diabetic retinopathy because VEGFR2 kinase activity, and VEGF-stimulated endothelial
(a) VEGF receptors are highly specific and (b) they are
cell proliferation in vitro. It has also been shown to
expressed in increased numbers during pathological vas-
inhibit VEGF-induced angiogenesis and to have antitumor
cular growth. Several different strategies have been found
activity in mice [143].
effective in preventing retinal neovascularization in mod-
els of ischemic retinopathy, including soluble VEGFR1,
anti-VEGF-neutralizing antibodies, soluble VEGF-receptor
chimeric proteins, antisense oligonucleotides, and inhibi- Inhibitors of VEGF expression
tion of a VEGF-specific protein kinase [49,63,131,132].
Two VEGF-directed therapies are currently under eval-
uation in clinical trials for use in the treatment of Knowledge of the transcriptional events associated with
angiogenesis in ocular diseases. EYE001 (macugen) is a VEGF expression in diabetic retinopathy is limited.
VEGF antisense pegylated RNA aptamer (oligonucleotide However, investigators at the National Cancer Institute
that acts like a high-affinity anti-VEGF antibody) that is have identified a number of small molecule inhibitors
now being tested for use in treating neovascularization in of HIF-1 activity and VEGF expression that are being
age-related macular degeneration and diabetic retinopa- developed as inhibitors of tumor angiogenesis [144].
thy [132]. Another therapy now being evaluated for use These molecules may also be useful for treatment of
in macular degeneration involves the use of a monoclonal diabetic retinopathy.
antibody fragment directed against VEGF [133]. Both of A number of inhibitors of hypoxia-inducible factor-
these agents are administered by intravitreous injection. 1 have been shown to negatively modulate VEGF
In addition to the above approaches, extensive clinical expression. This is the case of the immunosuppressant
research is being done to evaluate the effectiveness rapamycin that has been shown to possess an antian-
of VEGF inhibitors for preventing tumor angiogenesis. giogenic activity by inhibiting VEGF expression [145].
Phase II/III clinical trials are now in progress testing a Several studies addressing rapamycin effects on VEGF
recombinant human anti-VEGF antibody directed against expression have shown that it is a negative modula-
free VEGF [44,134,135]. This antibody functions by tor of the phosphatidylinositol 3 -kinase/Akt-dependent
binding to VEGF and blocking its ability to bind to mTOR (mammalian target of rapamycin) activation. This
its receptor. Another antibody directed against VEGFR2 leads to a reduction in the transactivating function of
is now in Phase I trials for treatment of cancer [44]. hypoxia-inducible factor-1 [146–148]. Recent studies
This monoclonal antibody has been shown to block both conducted with cultured retinal endothelial cells indicated
VEGFR-2-mediated permeability and tumor angiogenesis that the antioxidant N-(2-mercapto-2-methylpropionyl)-
in mice [136]. L-cysteine (bucillamine) inhibits VEGF expression by
An alternative approach has been the use of soluble inhibiting hypoxia-inducible factor-1 transcriptional activ-
VEGF receptors as a decoy to sequester VEGF and ity [149]. Another study in streptozotocin-induced dia-
reduce its bioavailablity. Recent studies using either betic rats showed that bucillamine-blocked diabetes-
adenovirus-mediated overexpression of soluble VEGFR1 induced increases in VEGF expression and vascular per-
[137] or liposomes carrying plasmids expressing soluble meability [150].
VEGFR1 [138] showed decreased tumor angiogenesis Less information is available regarding approaches to
and increased tumor apoptosis. Similarly, studies done inhibit STAT3 activation of VEGF expression. Because
in rats that had been implanted with tumor cells of the overexpression of VEGF and the angiogenic
infected with dominant negative expression of VEGFR- requirements for tumor growth, drug discovery pro-
2 showed impaired angiogenesis and reduced tumor grams are actively searching for peptide inhibitors of
vascular density [139]. STAT3 [151]. One such inhibitor has been identi-
Another technique that is now in Phase I/II clinical fied and will soon be available for laboratory research
trials for cancer therapy involves the use of ribozymes [152].

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
450 R. B. Caldwell et al.

HMG-CoA reductase inhibitors (statins) of prostenoid mediators of inflammation via the cyclooxy-
genase pathway [131,164]. High-dose aspirin (nonselec-
Recent studies have indicated that statins possess remark- tive inhibitor of cyclooxygenase-1 and cyclooxygenase-2)
able vasoprotective effects in a wide variety of pathologi- has been reported to reduce the incidence of diabetic
cal conditions including diabetes [153]. Statins’ protective retinopathy in patients [165] and to prevent the devel-
actions are exerted mainly on the microvasculature and opment of retinal vascular changes in diabetic dogs
appear to be independent of their lipid lowering proper- [166]. Because retinal expression of cyclooxygenase-2,
ties and rather to be the result of their antiinflammatory but not cyclooxygenase-1 is elevated during diabetes, it
and antioxidant properties as well as their actions as is considered likely that cyclooxygenase-2 activity may
inducers and agonists of nitric oxide synthase. Statins’ be responsible for the aspirin effect [167]. Studies in
pleiotropic effects are due in part to their molecular action the streptozotocin-induced diabetic rat model showing
as inhibitors of the HMG-CoA reductase, which catalyzes that cyclooxygenase-2 inhibitor treatment inhibits reti-
the synthesis of L-mevalonate. L-mevalonate is a precursor nal VEGF expression and vascular leakage support this
of cholesterol and isoprenoid intermediates such as farne- hypothesis [117]. Further work is needed to determine
syl pyrophosphate and geranyl–geranyl pyrophosphate. the effects of selective inhibitors of cyclooxygenase-2 in
Isoprenoids are important lipid moieties needed for the preventing diabetic retinopathy in patients.
posttranslational modification of a variety of small G pro-
teins (Ras-like proteins) such as Rho and Rac-1 [154].
Statin-mediated inhibition of Rho leads to upregulation Antioxidants
of endothelial nitric oxide synthase (NOS3) activation
of the phosphatidylinositol 3 -kinase/Akt pathway and The role of oxidative stress in diabetic vascular
increased nitric oxide production and statin-induced inhi- dysfunction and VEGF overexpression is well established.
bition of Rac-1 results in blockade of NAD(P)H oxidase Production of superoxide anion can result from various
activity [155]. Statins have also been shown to down- mechanisms active in diabetes, including activation
regulate VEGF expression induced by advanced glycation of the mitochondrial electron-transport chain, glucose
end products and to inhibit angiogenesis associated with auto-oxidation, protein glycation, increased glucose flux
tumors [156–158]. These contrasting angiogenic and through the polyol pathway, and prostanoid production
angiostatic properties have been explained by in vitro [22,168]. In this regard, normalization of glucose-
studies showing a biphasic, dose-dependent effect of stimulated superoxide production has been found to block
statins [158,159]. However, several studies in experimen- at least three independent pathways of hyperglycemia-
tal animals and in patients indicate that statins suppress induced vascular damage [22]. Studies showing that
pathological angiogenesis at clinically relevant doses that VEGF itself induces formation of superoxide anion in
have been shown to induce angiogenesis in models of cultured endothelial cells and that superoxide anion
acute ischemia [160,161]. formation is required for VEGF’s angiogenic actions
Finally, statins have been shown to inhibit angiotensin- [76] suggest that superoxide inhibitors/scavengers may
induced activation of STAT3 in vitro [160]. If this process also improve diabetic retinopathy by inhibiting the
has a role in VEGF overexpression and the associated VEGF intracellular signaling pathway. Furthermore,
alterations in vascular function and growth during antioxidants such as vitamin E have been shown to prevent
diabetes, statin therapy may be useful in treating diabetic some of the vascular dysfunctions associated with diabetes
retinopathy. Anecdotal evidence indicates that lipid in animal models [169]. In one study of diabetic patients
exudation associated with breakdown of the blood-retinal who had minimal or no retinopathy, treatment with high-
barrier is reduced in diabetic patients treated with statins dose vitamin E for four months was found to significantly
[162]. However, the number of patients studied was reverse diabetes-induced alterations in retinal blood flow
small. Randomized clinical trials are needed to evaluate [170].
the efficacy of statins as therapy for diabetic retinopathy. Another potential target for antioxidant therapy
Because the statins are well known drugs whose efficacy, is peroxynitrite. Diabetes- and high-glucose-induced
side effects and toxicity have been well characterized in vascular dysfunction has long been thought to involve
more than ten years of clinical use, studies determining the inactivation of endothelium-derived nitric oxide by its
their efficacy in preventing retinal neovascularization and combination with superoxide anion to form peroxynitrite.
diabetic retinopathy will be important. This concept has been supported by research showing
nitrotyrosine formation in the plasma of diabetic patients
[32] and in blood vessels and retinas from diabetic
Cyclooxygenase-2 inhibitors rats [30,171–173]. Moreover, studies in diabetic rats
have also shown that increases in retinal nitrotyrosine
Retinas of diabetic rats as well as diabetic patients exhibit levels and lipid peroxidation are correlated with VEGF
inflammation associated alterations, such as vasodilata- overexpression and breakdown of the blood-retinal
tion, vascular leakage, platelet aggregation and leukocyte barrier and that all of these alterations can be prevented
adhesion [163]. High glucose treatment of cultured retinal by treatment with the peroxynitrite scavenger uric acid
endothelial cells has been shown to induce the production or the nitric oxide synthase inhibitor L-NAME [30].

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
VEGF and Diabetic Retinopathy 451

Preclinical studies in the mouse model for oxygen- Acknowledgements


induced retinopathy provide further support for the role
of peroxynitrite in retinal neovascular disease. These NIH-R01-EY04618, NIH-R01-EY11766, American Heart Associ-
studies have shown that increased peroxynitrite formation ation, and unrestricted research award to the Department of
is associated with retinal capillary dropout and that Ophthalmology from Research to Prevent Blindness
NOS3 deletion or treatment with a nitric oxide synthase
inhibitor diminishes VEGF expression and inhibits retinal
neovascularization [174,175]. References
1. Klein R, Klein BEK (eds). Vision disorders in diabetes. In:
Harris MI ed., Diabetes in America, 2nd edn, Bethesda, MD:
Conclusion National Institutes of Health, National Institute of Diabetes
and Digestive and Kidney Diseases. US Public Health Service
1995: (NIH Publ. no. 95–1468).
For many years, diabetic retinopathy has been the major 2. Klein R, Klein BE, Moss SE, Davis MD, DeMets DL. The
cause of new adult blindness in the industrialized world. Wisconsin epidemiologic study of diabetic retinopathy. II.
Recent improvements in the care and management Prevalence and risk of diabetic retinopathy when age at
diagnosis is less than 30 years. Arch Ophthalmol 1984; 102:
of diabetic patients have decreased the incidence and 520–526.
severity of diabetes complications in the retina as 3. Sjolie AK, Stephenson J, Aldington S, et al. Retinopathy and
well as other organ systems and advances in the vision loss in insulin-dependent diabetes in Europe. The
EURODIAB IDDM complications study. Ophthalmology 1997;
use of laser photocoagulation therapy have improved 104: 252–260.
the visual outcome for many of the patients that 4. The Diabetes Control and Complications Trial Research
do develop diabetic retinopathy. Nevertheless, many Group. The effect of intensive treatment of diabetes on the
development and progression of long-term complications in
patients still develop diabetic retinopathy in spite of insulin-dependent diabetes mellitus. N Engl J Med 1993; 329:
good diabetes control. Moreover, laser therapy maintains 977–986.
vision rather than recovering lost vision and can 5. Lu M, Kuroki M, Amano S, et al. Advanced glycation end
products increase retinal vascular endothelial growth factor
itself lead to additional visual loss. Therefore, there expression. J Clin Invest 1998; 101: 1219–1224.
is a great need for new therapies that are safe and 6. Klein R, Klein BE. Diabetic eye disease. Lancet 1997; 350:
effective for preventing and treating diabetes retinal 197–204.
7. Aiello LP, Gardner TW, King GL, et al. Diabetic retinopathy.
complications. Diabetes Care 1998; 21: 143–156.
A number of promising therapies are currently under 8. Frank RN. Potential new medical therapies for diabetic
investigation in clinical trials, including inhibitors of retinopathy: protein kinase C inhibitors. Am J Ophthalmol
2002; 133: 693–698.
angiotensin converting enzyme activity, angiotensin 9. UK Prospective Diabetes Study Group. Tight blood pressure
receptor blockers and inhibitors of the protein kinase control and risk of macrovascular and microvascular
C beta isoform. Recently, a wealth of clinical and complications in type 2 diabetes: UKPDS 38. BMJ 1998; 317:
703–713.
experimental evidence has accumulated to demonstrate 10. Sheetz MJ, King GL. Molecular understanding of hyper-
that overexpression of VEGF has a key role in diabetes- glycemia’s adverse effects for diabetic complications. Jama
induced retinal vascular dysfunction. Therefore, the 2002; 288: 2579–2588.
11. Aiello LP. The Potential role of PKC beta in diabetic retinopathy
development of agents that directly target VEGF and and macular edema. Surv Ophthalmol 2002; 47(Suppl. 2):
its receptors is an area of active clinical research. S263–S269.
New therapeutic strategies not yet in clinical trials 12. Hagedorn M, Bikfalvi A. Target molecules for anti-angiogenic
therapy: from basic research to clinical trials. Crit Rev Oncol
but supported by promising preclinical data involve Hematol 2000; 34: 89–110.
the use of pharmacological approaches to block VEGF 13. Novak K. American Association for Cancer Research.
overexpression or prevent its adverse effects on vascular Angiogenesis inhibitors revised and revived at AACR. Nat Med
2002; 8: 427.
permeability and growth. These include various inhibitors 14. Casey R, Li WW. Factors controlling ocular angiogenesis. Am J
of VEGF transcriptional activation, the statin HMG- Ophthalmol 1997; 124: 521–529.
CoA reductase inhibitors, cyclooxygenase-2 inhibitors and 15. Duh E, Aiello LP. Vascular endothelial growth factor and
diabetes: the agonist versus antagonist paradox. Diabetes 1999;
antioxidant therapies. 48: 1899–1906.
A new area that is just beginning to be considered 16. Ciardella AP, Donsoff IM, Guyer DR, Adamis AP, Yannuzzi LA.
for potential use in the treatment of diabetes-induced Antiangiogenesis agents. Ophthalmol Clin North Am 2002; 15:
453–458.
vascular disease involves the use of endothelial progenitor 17. Chakrabarti S, Cukiernik M, Hileeto D, Evans T, Chen S. Role
cells. While the specific role of endothelial progenitor of vasoactive factors in the pathogenesis of early changes
cells in diabetic retinopathy is not yet clear, the in diabetic retinopathy. Diabetes Metab Res Rev 2000; 16:
393–407.
results of initial studies in other models of ischemic 18. Sanchez-Elsner T, Botella LM, Velasco B, Corbi A, Attisano L,
retinopathy have supported their role in pathological Bernabeu C. Synergistic cooperation between hypoxia and
angiogenesis and suggested a potential use as tools for transforming growth factor-beta pathways on human vascular
endothelial growth factor gene expression. J Biol Chem 2001;
drug delivery in the treatment of retinal vascular disease. 276: 38 527–38 535.
Further investigation is needed to establish the role 19. Miller JW, Adamis AP, Aiello LP. Vascular endothelial growth
of endothelial progenitor cells in ischemic retinopathy factor in ocular neovascularization and proliferative diabetic
retinopathy. Diabetes Metab Rev 1997; 13: 37–50.
and to determine their involvement in diabetes retinal 20. Hammes HP, Lin J, Bretzel RG, Brownlee M, Breier G.
complications. Upregulation of the vascular endothelial growth

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
452 R. B. Caldwell et al.

factor/vascular endothelial growth factor receptor system in 41. Patel JM, Abeles AJ, Block ER. Nitric oxide exposure and
experimental background diabetic retinopathy of the rat. sulfhydryl modulation alter L-arginine transport in cultured
Diabetes 1998; 47: 401–406. pulmonary artery endothelial cells. Free Radic Biol Med 1996;
21. Gilbert RE, Vranes D, Berka JL, et al. Vascular endothelial 20: 629–637.
growth factor and its receptors in control and diabetic rat 42. El-Remessy A, Bartoli M, Fulton D, Caldwell RW, Caldwell RB.
eyes. Lab Invest 1998; 78: 1017–1027. Oxidative stress inactivates the VEGF survival pathway in
22. Brownlee M. Biochemistry and molecular cell biology of endothelial cells exposed to high glucose. Circ 2002; 106:
diabetic complications. Nature 2001; 414: 813–820. 281.
23. Kowluru RA, Tang J, Kern TS. Abnormalities of retinal 43. Zou MH, Shi C, Cohen RA. High glucose via peroxynitrite
metabolism in diabetes and experimental galactosemia. VII. causes tyrosine nitration and inactivation of prostacyclin
Effect of long-term administration of antioxidants on the synthase that is associated with thromboxane/prostaglandin
development of retinopathy. Diabetes 2001; 50: 1938–1942. H(2) receptor-mediated apoptosis and adhesion molecule
24. Kowluru RA, Engerman RL, Kern TS. Abnormalities of retinal expression in cultured human aortic endothelial cells. Diabetes
metabolism in diabetes or experimental galactosemia VIII. 2002; 51: 198–203.
Prevention by aminoguanidine. Curr Eye Res 2000; 21: 44. McMahon G. VEGF receptor signaling in tumor angiogenesis.
814–819. Oncologist 2000; 5: 3–10.
25. Ellis EA, Guberski DL, Somogyi-Mann M, Grant MB. Increased 45. Ferrara N. Role of vascular endothelial growth factor in
H2O2, vascular endothelial growth factor and receptors in the regulation of physiological angiogenesis. Am J Physiol Cell
retina of the BBZ/Wor diabetic rat. Free Radic Biol Med 2000; Physiol 2001; 280: C1358–C1366.
28: 91–101. 46. Ku DD, Zaleski JK, Liu S, Brock TA. Vascular endothelial
26. Ellis EA, Grant MB, Murray FT, et al. Increased NADH oxidase growth factor induces EDRF-dependent relaxation in coronary
activity in the retina of the BBZ/Wor diabetic rat. Free Radic arteries. Am J Physiol 1993; 265: H586–H592.
Biol Med 1998; 24: 111–120. 47. Alon T, Hemo I, Itin A, Pe’er J, Stone J, Keshet E. Vascular
27. Kuroki M, Voest EE, Amano S, et al. Reactive oxygen endothelial growth factor acts as a survival factor for newly
intermediates increase vascular endothelial growth factor formed retinal vessels and has implications for retinopathy of
expression in vitro and in vivo. J Clin Invest 1996; 98: prematurity. Nat Med 1995; 1: 1024–1028.
1667–1675. 48. Borgstrom P, Gold DP, Hillan KJ, Ferrara N. Importance of
28. Obrosova IG, Minchenko AG, Marinescu V, et al. Antioxidants VEGF for breast cancer angiogenesis in vivo: implications
attenuate early up regulation of retinal vascular endothelial from intravital microscopy of combination treatments with an
growth factor in streptozotocin-diabetic rats. Diabetologia anti-VEGF neutralizing monoclonal antibody and doxorubicin.
2001; 44: 1102–1110. Anticancer Res 1999; 19: 4203–4214.
29. Du Y, Smith MA, Miller CM, Kern TS. Diabetes-induced 49. Aiello LP, Pierce EA, Foley ED, et al. Suppression of retinal
nitrative stress in the retina, and correction by aminoguanidine. neovascularization in vivo by inhibition of vascular endothelial
J Neurochem 2002; 80: 771–779. growth factor (VEGF) using soluble VEGF-receptor chimeric
30. El-Remessy AB, Behzadian MA, Abou-Mohamed G, Franklin T, proteins. Proc Natl Acad Sci U S A 1995; 92: 10 457–10 461.
Caldwell RW, Caldwell RB. Experimental diabetes causes
50. Simorre-Pinatel V, Guerrin M, Chollet P, et al. Vasculotropin-
breakdown of the blood-retina barrier by a mechanism
VEGF stimulates retinal capillary endothelial cells through
involving tyrosine nitration and increases in expression of
an autocrine pathway. Invest Ophthalmol Vis Sci 1994; 35:
vascular endothelial growth factor and urokinase plasminogen
3393–3400.
activator receptor. Am J Pathol 2003; 162: 1995–2004.
51. Adamis AP, Shima DT, Yeo KT, et al. Synthesis and secretion of
31. Kossenjans W, Eis A, Sahay R, Brockman D, Myatt L. Role of
vascular permeability factor/vascular endothelial growth factor
peroxynitrite in altered fetal-placental vascular reactivity in
by human retinal pigment epithelial cells. Biochem Biophys Res
diabetes or preeclampsia. Am J Physiol Heart Circ Physiol 2000;
Commun 1993; 193: 631–638.
278: H1311–H1319.
32. Ceriello A, Mercuri F, Quagliaro L, et al. Detection of 52. Behzadian MA, Wang XL, Shabrawey M, Caldwell RB. Effects
nitrotyrosine in the diabetic plasma: evidence of oxidative of hypoxia on glial cell expression of angiogenesis-regulating
stress. Diabetologia 2001; 44: 834–838. factors VEGF and TGF-beta. Glia 1998; 24: 216–225.
33. Graier WF, Simecek S, Kukovetz WR, Kostner GM. High 53. Brooks SE, Gu X, Kaufmann PM, Marcus DM, Caldwell RB.
D-glucose-induced changes in endothelial Ca2+/EDRF Modulation of VEGF production by pH and glucose in retinal
signaling are due to generation of superoxide anions. Diabetes Muller cells. Curr Eye Res 1998; 17: 875–882.
1996; 45: 1386–1395. 54. Cheng T, Cao W, Wen R, Steinberg RH, LaVail MM.
34. Graier WF, Posch K, Wascher TC, Kostner GM. Role of Prostaglandin E2 induces vascular endothelial growth factor
superoxide anions in changes of endothelial vasoactive and basic fibroblast growth factor mRNA expression in cultured
response during acute hyperglycemia. Horm Metab Res 1997; rat Muller cells. Invest Ophthalmol Vis Sci 1998; 39: 581–591.
29: 622–626. 55. Hata Y, Clermont A, Yamauchi T, et al. Retinal expression,
35. Graier WF, Posch K, Fleischhacker E, Wascher TC, Kostner GM. regulation, and functional bioactivity of prostacyclin-
Increased superoxide anion formation in endothelial cells stimulating factor. J Clin Invest 2000; 106: 541–550.
during hyperglycemia: an adaptive response or initial step 56. Nie D, Lamberti M, Zacharek A, et al. Thromboxane A(2)
of vascular dysfunction? Diabetes Res Clin Pract 1999; 45: regulation of endothelial cell migration, angiogenesis, and
153–160. tumor metastasis. Biochem Biophys Res Commun 2000; 267:
36. El-Remessy A, Abou-Mohamed G, Caldwell RW, Caldwell RB. 245–251.
High glucose increases tyrosine nitration and superoxide anion 57. Behzadian MA, El-Remessy AB, Franklin T, Caldwell RB.
formation in endothelial cells: role of eNOS uncoupling and Mechanism of glucose induced urokinase receptor (uPAR)
aldose reductase activation. Invest Ophthalmol Vis Sci 2003; expression in retinal endothelial cells, possible role of ROS
44: 3135–3143. and peroxynitrite formation. Invest Ophthalmol Vis Sci 2003;
37. Salgo MG, Bermudez E, Squadrito GL, Pryor WA. Peroxynitrite 44: Abstract 3905.
causes DNA damage and oxidation of thiols in rat thymocytes 58. Qaum T, Xu Q, Joussen AM, et al. VEGF-initiated blood-retinal
[corrected]. [published erratum appears in Arch Biochem barrier breakdown in early diabetes. Invest Ophthalmol Vis Sci
Biophys 1995; 324(1): 200]; Arch Biochem Biophys 1995; 322: 2001; 42: 2408–2413.
500–505. 59. Dorey CK, Aouididi S, Reynaud X, Dvorak HF, Brown LF.
38. Salgo MG, Squadrito GL, Pryor WA. Peroxynitrite causes Correlation of vascular permeability factor/vascular
apoptosis in rat thymocytes. Biochem Biophys Res Commun endothelial growth factor with extraretinal neovascularization
1995; 215: 1111–1118. in the rat. Arch Ophthalmol 1996; 114: 1210–1217.
39. Zhuang S, Simon G. Peroxynitrite-induced apoptosis involves 60. Donahue ML, Phelps DL, Watkins RH, LoMonaco MB,
activation of multiple caspases in HL-60 cells. Am J Physiol Cell Horowitz S. Retinal vascular endothelial growth factor (VEGF)
Physiol 2000; 279: C341–C351. mRNA expression is altered in relation to neovascularization in
40. Milstien S, Katusic Z. Oxidation of tetrahydrobiopterin by oxygen induced retinopathy. Curr Eye Res 1996; 15: 175–184.
peroxynitrite: implications for vascular endothelial function. 61. Robbins SG, Conaway JR, Ford BL, Roberto KA, Penn JS.
Biochem Biophys Res Commun 1999; 263: 681–684. Detection of vascular endothelial growth factor (VEGF) protein

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
VEGF and Diabetic Retinopathy 453

in vascular and non-vascular cells of the normal and oxygen- 84. Schaefer LK, Ren Z, Fuller GN, Schaefer TS. Constitutive
injured rat retina. Growth Factors 1997; 14: 229–241. activation of Stat3alpha in brain tumors: localization to tumor
62. Robinson GS, Pierce EA, Rook SL, Foley E, Webb R, Smith LE. endothelial cells and activation by the endothelial tyrosine
Oligodeoxynucleotides inhibit retinal neovascularization in a kinase receptor (VEGFR-2). Oncogene 2002; 21: 2058–2065.
murine model of proliferative retinopathy. Proc Natl Acad Sci 85. Valdembri D, Serini G, Vacca A, Ribatti D, Bussolino F. In vivo
U S A 1996; 93: 4851–4856. activation of JAK2/STAT-3 pathway during angiogenesis
63. Ozaki H, Seo MS, Ozaki K, et al. Blockade of vascular induced by GM-CSF. FASEB J 2002; 16: 225–227.
endothelial cell growth factor receptor signaling is sufficient 86. Bartoli M, Gu X, Tsai NT, et al. Vascular endothelial growth
to completely prevent retinal neovascularization. Am J Pathol factor activates STAT proteins in aortic endothelial cells. J Biol
2000; 156: 697–707. Chem 2000; 275: 33 189–33 192.
64. Carmeliet P, Ferreira V, Breier G, et al. Abnormal blood vessel 87. Bartoli M, Platt D, Lemtalsi T, et al. VEGF differentially
development and lethality in embryos lacking a single VEGF activates STAT3 in microvascular endothelial cells. FASEB J
allele. Nature 1996; 380: 435–439. 2003; 11: 1562–1564.
65. Levy AP, Levy NS, Goldberg MA. Post-transcriptional regula- 88. Platt DH, Bartoli M, El-Remessy AB, Al-Shabrawey M, Mar-
tion of vascular endothelial growth factor by hypoxia. J Biol rero MB, Caldwell RB. Peroxynitrite induces transcriptional
Chem 1996; 271: 2746–2753. activation of vascular endothelial growth factor in bovine
66. Forsythe JA, Jiang BH, Iyer NV, et al. Activation of vascular retinal endothelial cells. Invest Ophthalmol Vis Sci 2003; 44:
endothelial growth factor gene transcription by hypoxia- Abstract 2097.
inducible factor 1. Mol Cell Biol 1996; 16: 4604–4613. 89. De Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N,
67. Wang GL, Semenza GL. Purification and characterization of Williams LT. The fms-like tyrosine kinase, a receptor for
hypoxia-inducible factor 1. J Biol Chem 1995; 270: 1230–1237. vascular endothelial growth factor. Science 1992; 255:
68. Wenger RH. Cellular adaptation to hypoxia: O2-sensing protein 989–991.
hydroxylases, hypoxia-inducible transcription factors, and O2- 90. Seetharam L, Gotoh N, Maru Y, Neufeld G, Yamaguchi S,
regulated gene expression. FASEB J 2002; 16: 1151–1162. Shibuya M. A unique signal transduction from FLT tyrosine
69. Poulaki V, Qin W, Joussen AM, et al. Acute intensive insulin kinase, a receptor for vascular endothelial growth factor VEGF.
therapy exacerbates diabetic blood-retinal barrier breakdown Oncogene 1995; 10: 135–147.
via hypoxia-inducible factor-1alpha and VEGF. J Clin Invest 91. Terman BI, Carrion ME, Kovacs E, Rasmussen BA, Eddy RL,
2002; 109: 805–815. Shows TB. Identification of a new endothelial cell growth factor
70. Ryuto M, Ono M, Izumi H, et al. Induction of vascular receptor tyrosine kinase. Oncogene 1991; 6: 1677–1683.
endothelial growth factor by tumor necrosis factor alpha in 92. Terman BI, Dougher-Vermazen M, Carrion ME, et al. Identifi-
human glioma cells. Possible roles of SP-1. J Biol Chem 1996; cation of the KDR tyrosine kinase as a receptor for vascular
271: 28 220–28 228. endothelial cell growth factor. Biochem Biophys Res Commun
71. Li J, Perrella MA, Tsai JC, et al. Induction of vascular 1992; 187: 1579–1586.
endothelial growth factor gene expression by interleukin-1 93. Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role of the
beta in rat aortic smooth muscle cells. J Biol Chem 1995; 270: Flt-1 receptor tyrosine kinase in regulating the assembly of
308–312.
vascular endothelium. Nature 1995; 376: 66–70.
72. Pertovaara L, Kaipainen A, Mustonen T, et al. Vascular
94. Shalaby F, Rossant J, Yamaguchi TP, et al. Failure of blood-
endothelial growth factor is induced in response to
island formation and vasculogenesis in Flk-1-deficient mice.
transforming growth factor-beta in fibroblastic and epithelial
Nature 1995; 376: 62–66.
cells. J Biol Chem 1994; 269: 6271–6274.
95. Fong GH, Zhang L, Bryce DM, Peng J. Increased hemangioblast
73. Goad DL, Rubin J, Wang H, Tashjian AH Jr, Patterson C.
commitment, not vascular disorganization, is the primary
Enhanced expression of vascular endothelial growth factor
defect in flt-1 knock-out mice. Development 1999; 126:
in human SaOS-2 osteoblast-like cells and murine osteoblasts
3015–3025.
induced by insulin-like growth factor I. Endocrinology 1996;
137: 2262–2268. 96. Grant MB, May WS, Caballero S, et al. Adult hematopoietic
74. Cohen T, Nahari D, Cerem LW, Neufeld G, Levi BZ. Interleukin stem cells provide functional hemangioblast activity during
6 induces the expression of vascular endothelial growth factor. retinal neovascularization. Nat Med 2002; 8: 607–612.
J Biol Chem 1996; 271: 736–741. 97. Luttun A, Tjwa M, Moons L, et al. Revascularization of
75. Hoffmann S, Friedrichs U, Eichler W, Rosenthal A, Wiede- ischemic tissues by PlGF treatment, and inhibition of tumor
mann P. Advanced glycation end products induce choroidal angiogenesis, arthritis and atherosclerosis by anti-Flt1. Nat
endothelial cell proliferation, matrix metalloproteinase-2 and Med 2002; 8: 831–840.
VEGF upregulation in vitro. Graefes Arch Clin Exp Ophthalmol 98. Zachary I, Gliki G. Signaling transduction mechanisms
2002; 240: 996–1002. mediating biological actions of the vascular endothelial growth
76. Ushio-Fukai M, Tang Y, Fukai T, et al. Novel role of factor family. Cardiovasc Res 2001; 49: 568–581.
gp91(phox)-containing NAD(P)H oxidase in vascular 99. Witmer AN, Blaauwgeers HG, Weich HA, Alitalo K, Vrensen
endothelial growth factor-induced signaling and angiogenesis. GF, Schlingemann RO. Altered expression patterns of VEGF
Circ Res 2002; 91: 1160–1167. receptors in human diabetic retina and in experimental VEGF-
77. Platt DH, Bartoli M, El-Remessy AB, Al-Shabrawey M, Mar- induced retinopathy in monkey. Invest Ophthalmol Vis Sci 2002;
rero MB, Caldwell RB. Peroxynitrite induces transcriptional 43: 849–857.
activation of vascular endothelial growth factor in bovine 100. Antonetti DA, Barber AJ, Khin S, Lieth E, Tarbell JM,
retinal endothelial cells. Invest Ophthalmol Vis Sci 2003; 44: Gardner TW. Penn State Retina Research Group. Vascular
Abstract 2097. permeability in experimental diabetes is associated with
78. Ihle JN. The Stat family in cytokine signaling. Curr Opin Cell reduced endothelial occludin content: vascular endothelial
Biol 2001; 13: 211–217. growth factor decreases occludin in retinal endothelial cells.
79. Mui AL. The role of STATs in proliferation, differentiation, and Diabetes 1998; 47: 1953–1959.
apoptosis. Cell Mol Life Sci 1999; 55: 1547–1558. 101. Adamis AP, Miller JW, Bernal M-T, et al. Increased vascular
80. Jove R. Preface: STAT signaling. Oncogene 2000; 19: endothelial growth factor levels in the vitreous of eyes with
2466–2467. proliferative diabetic retinopathy. Am J Ophthalmol 1994; 118:
81. Niu G, Wright KL, Huang M, et al. Constitutive Stat3 activity 445–450.
up-regulates VEGF expression and tumor angiogenesis. 102. Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial
Oncogene 2002; 21: 2000–2008. growth factor in ocular fluid of patients with diabetic
82. Funamoto M, Fujio Y, Kunisada K, et al. Signal transducer and retinopathy and other retinal disorders. New Eng J Med 1994;
activator of transcription 3 is required for glycoprotein 130- 331: 1480–1487.
mediated induction of vascular endothelial growth factor in 103. Takeda M, Mori F, Yoshida A, et al. Constitutive nitric oxide
cardiac myocytes. J Biol Chem 2000; 275: 10 561–10 566. synthase is associated with retinal vascular permeability in
83. Osugi T, Oshima Y, Fujio Y, et al. Cardiac-specific activation of early diabetic rats. Diabetologia 2001; 44: 1043–1050.
signal transducer and activator of transcription 3 promotes 104. Joussen AM, Poulaki V, Tsujikawa A, et al. Suppression of
vascular formation in the heart. J Biol Chem 2002; 277: diabetic retinopathy with angiopoietin-1. Am J Pathol 2002;
6676–6681. 160: 1683–1693.

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
454 R. B. Caldwell et al.

105. Senger DR, Galli SJ, Dvorak AM, Perruzzi CA, Harvey VS, 125. Rousseau S, Houle F, Landry J, Huot J. p38 MAP kinase
Dvorak HF. Tumor cells secrete a vascular permeability factor activation by vascular endothelial growth factor mediates actin
that promotes accumulation of ascites fluid. Science 1983; 219: reorganization and cell migration in human endothelial cells.
983–985. Oncogene 1997; 15: 2169–2177.
106. Feng Y, Venema VJ, Venema RC, Tsai N, Behzadian MA, 126. Gratton JP, Morales-Ruiz M, Kureishi Y, Fulton D, Walsh K,
Caldwell RB. VEGF-induced permeability increase is mediated Sessa WC. Akt down-regulation of p38 signaling provides
by caveolae. Invest Ophthalmol Vis Sci 1999; 40: 157–167. a novel mechanism of vascular endothelial growth factor-
107. Behzadian MA, Windsor LJ, Ghaly N, Liou G, Tsai NT, mediated cytoprotection in endothelial cells. J Biol Chem 2001;
Caldwell RB. VEGF-induced paracellular permeability in 276: 30 359–30 365.
cultured endothelial cells involves urokinase and its receptor. 127. Gu X, El-Remessy AB, Brooks SE, Al-Shabrawey M, Tsai NT,
FASEB J 2003; 19: 19. Caldwell RB. Hyperoxia induces retinal vascular endothelial
108. Mandriota SJ, Seghezzi G, Vassalli JD, et al. Vascular cell apoptosis through formation of peroxynitrite. Am J Physiol
endothelial growth factor increases urokinase receptor Cell Physiol 2003; 285: C546–C554.
expression in vascular endothelial cells. J Biol Chem 1995; 128. Hellberg CB, Boggs SE, Lapetina EG. Phosphatidylinositol
270: 9709–9716. 3-kinase is a target for protein tyrosine nitration. Biochem
109. Barber AJ, Antonetti DA, Gardner TW. The Penn State Retina Biophys Res Commun 1998; 252: 313–317.
Research Group. Altered expression of retinal occludin and 129. Misko TP, Highkin MK, Veenhuizen AW, et al. Characterization
glial fibrillary acidic protein in experimental diabetes. Invest of the cytoprotective action of peroxynitrite decomposition
Ophthalmol Vis Sci 2000; 41: 3561–3568. catalysts. J Biol Chem 1998; 273: 15 646–15 653.
110. He H, Venema VJ, Gu X, Venema RC, Marrero MB, 130. Salvemini D, Wang ZQ, Stern MK, Currie MG, Misko TP.
Caldwell RB. Vascular endothelial growth factor signals Peroxynitrite decomposition catalysts: therapeutics for
endothelial cell production of nitric oxide and prostacyclin peroxynitrite-mediated pathology. Proc Natl Acad Sci U S A
through flk-1/KDR activation of c-Src. J Biol Chem 1999; 274: 1998; 95: 2659–2663.
25 130–25 135. 131. Sone H, Kawakami Y, Segawa T, et al. Effects of intraocular
111. Ferrara N. Molecular and biological properties of vascular or systemic administration of neutralizing antibody against
endothelial growth factor. J Mol Med 1999; 77: 527–543. vascular endothelial growth factor on the murine experimental
112. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative model of retinopathy. Life Sci 1999; 65: 2573–2580.
progenitor endothelial cells for angiogenesis. Science 1997; 132. Eyetech Study Group. Preclinical and phase 1A clinical
275: 964–967. evaluation of an anti-VEGF pegylated aptamer (EYE001) for
113. Schatteman GC, Hanlon HD, Jiao C, Dodds SG, Christy BA. the treatment of exudative age-related macular degeneration.
Blood-derived angioblasts accelerate blood-flow restoration in Retina 2002; 22: 143–152.
diabetic mice. J Clin Invest 2000; 106: 571–578. 133. Krzystolik MG, Afshari MA, Adamis AP, et al. Prevention of
114. Tepper OM, Galiano RD, Capla JM, et al. Human endothelial experimental choroidal neovascularization with intravitreal
progenitor cells from type II diabetics exhibit impaired anti-vascular endothelial growth factor antibody fragment.
proliferation, adhesion, and incorporation into vascular Arch Ophthalmol 2002; 120: 338–346.
134. Chen HX, Gore-Langton RE, Cheson BD. Clinical trials referral
structures. Circulation 2002; 106: 2781–2786.
resource: current clinical trials of the anti-VEGF monoclonal
115. Otani A, Kinder K, Ewalt K, Otero FJ, Schimmel P, Friedlan-
antibody bevacizumab. Oncology (Huntingt) 2001; 15: 1017,
der M. Bone marrow-derived stem cells target retinal astrocytes
1020, 1023–1016.
and can promote or inhibit retinal angiogenesis. Nat Med 2002;
135. Gordon MS, Margolin K, Talpaz M, et al. Phase I safety and
8: 1004–1010.
pharmacokinetic study of recombinant human anti-vascular
116. Yamagishi S, Yonekura H, Yamamoto Y, et al. Advanced
endothelial growth factor in patients with advanced cancer. J
glycation end products-driven angiogenesis in vitro. Induction
Clin Oncol 2001; 19: 843–850.
of the growth and tube formation of human microvascular
136. Brekken RA, Overholser JP, Stastny VA, Waltenberger J,
endothelial cells through autocrine vascular endothelial growth Minna JD, Thorpe PE. Selective inhibition of vascular
factor. J Biol Chem 1997; 272: 8723–8730. endothelial growth factor (VEGF) receptor 2 (KDR/Flk-1)
117. Ayalasomayajula SP, Kompella UB. Celecoxib, a selective activity by a monoclonal anti-VEGF antibody blocks tumor
cyclooxygenase-2 inhibitor, inhibits retinal vascular endothelial growth in mice. Cancer Res 2000; 60: 5117–5124.
growth factor expression and vascular leakage in a 137. Takayama K, Ueno H, Nakanishi Y, et al. Suppression of tumor
streptozotocin-induced diabetic rat model. Eur J Pharmacol angiogenesis and growth by gene transfer of a soluble form
2003; 458: 283–289. of vascular endothelial growth factor receptor into a remote
118. Joussen AM, Poulaki V, Mitsiades N, et al. Nonsteroidal anti- organ. Cancer Res 2000; 60: 2169–2177.
inflammatory drugs prevent early diabetic retinopathy via 138. Mori A, Arii S, Furutani M, et al. Soluble Flt-1 gene therapy for
TNF-alpha suppression. FASEB J 2002; 16: 438–440. peritoneal metastases using HVJ-cationic liposomes. Gene Ther
119. Joussen AM, Poulaki V, Qin W, et al. Retinal vascular 2000; 7: 1027–1033.
endothelial growth factor induces intercellular adhesion 139. Machein MR, Risau W, Plate KH. Antiangiogenic gene therapy
molecule-1 and endothelial nitric oxide synthase expression in a rat glioma model using a dominant-negative vascular
and initiates early diabetic retinal leukocyte adhesion in vivo. endothelial growth factor receptor 2. Hum Gene Ther 1999; 10:
Am J Pathol 2002; 160: 501–509. 1117–1128.
120. Funatsu H, Yamashita H, Noma H, et al. Outcome of vitreous 140. Weng DE, Usman N. Angiozyme: a novel angiogenesis
surgery and the balance between vascular endothelial growth inhibitor. Curr Oncol Rep 2001; 3: 141–146.
factor and endostatin. Invest Ophthalmol Vis Sci 2003; 44: 141. Vajkoczy P, Menger MD, Vollmar B, et al. Inhibition of tumor
1042–1047. growth, angiogenesis, and microcirculation by the novel Flk-1
121. Takahashi K, Saishin Y, Silva RL, et al. Intraocular expression inhibitor SU5416 as assessed by intravital multi-fluorescence
of endostatin reduces VEGF-induced retinal vascular videomicroscopy. Neoplasia 1999; 1: 31–41.
permeability, neovascularization, and retinal detachment. 142. Zhu Z, Witte L. Inhibition of tumor growth and metastasis by
FASEB J 2003; 28: 28. targeting tumor-associated angiogenesis with antagonists to
122. Mizutani M, Kern TS, Lorenzi M. Accelerated death of retinal the receptors of vascular endothelial growth factor. Invest New
microvascular cells in human and experimental diabetic Drugs 1999; 17: 195–212.
retinopathy. J Clin Invest 1996; 97: 2883–2890. 143. Wedge SR, Ogilvie DJ, Dukes M, et al. ZD4190: an orally
123. Gerber HP, Dixit V, Ferrara N. Vascular endothelial growth active inhibitor of vascular endothelial growth factor signaling
factor induces expression of the antiapoptotic proteins Bcl-2 with broad-spectrum antitumor efficacy. Cancer Res 2000; 60:
and A1 in vascular endothelial cells. J Biol Chem 1998; 273: 970–975.
13 313–13 316. 144. Rapisarda A, Uranchimeg B, Scudiero DA, et al. Identification
124. Gerber HP, McMurtrey A, Kowalski J, et al. Vascular of small molecule inhibitors of hypoxia-inducible factor 1
endothelial growth factor regulates endothelial cell transcriptional activation pathway. Cancer Res 2002; 62:
survival through the phosphatidylinositol 3 -kinase/Akt signal 4316–4324.
transduction pathway. Requirement for Flk-1/KDR activation. 145. Guba M, von Breitenbuch P, Steinbauer M, et al. Rapamycin
J Biol Chem 1998; 273: 30 336–30 343. inhibits primary and metastatic tumor growth by

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.
VEGF and Diabetic Retinopathy 455

antiangiogenesis: involvement of vascular endothelial growth angiotensin II type 1 receptor blocker, valsartan, on vascular
factor. Nat Med 2002; 8: 128–135. neointimal formation. Circulation 2003; 107: 106–112.
146. Hudson CC, Liu M, Chiang GG, et al. Regulation of hypoxia- 161. Kawata S, Yamasaki E, Nagase T, et al. Effect of pravastatin on
inducible factor 1alpha expression and function by the survival in patients with advanced hepatocellular carcinoma. A
mammalian target of rapamycin. Mol Cell Biol 2002; 22: randomized controlled trial. Br J Cancer 2001; 84: 886–891.
7004–7014. 162. Chowdhury TA, Hopkins D, Dodson PM, Vafidis GC. The role
147. Treins C, Giorgetti-Peraldi S, Murdaca J, Semenza GL, Van of serum lipids in exudative diabetic maculopathy: is there a
Obberghen E. Insulin stimulates hypoxia-inducible factor 1 place for lipid lowering therapy? Eye 2002; 16: 689–693.
through a phosphatidylinositol 3-kinase/target of rapamycin- 163. Adamis AP. Is diabetic retinopathy an inflammatory disease?
dependent signaling pathway. J Biol Chem 2002; 277: Br J Ophthalmol 2002; 86: 363–365.
27 975–27 981. 164. Sone H, Okuda Y, Kawakami Y, Yamashita K. Effects of high
148. Zhong H, Chiles K, Feldser D, et al. Modulation of hypoxia- glucose concentration and a thromboxane synthase inhibitor
inducible factor 1alpha expression by the epidermal on the production of thromboxane A2 and prostaglandin I2
growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP and E2 by retinal endothelial cells. Life Sci 1996; 58: 239–243.
pathway in human prostate cancer cells: implications for 165. Powell ED, Field RA. Studies on salicylates and complement in
tumor angiogenesis and therapeutics. Cancer Res 2000; 60: diabetes. Diabetes 1966; 15: 730–733.
1541–1545. 166. Kern TS, Engerman RL. Pharmacological inhibition of diabetic
149. Koyama S, Takagi H, Otani A, Oh H, Nishimura K, Honda Y. retinopathy: aminoguanidine and aspirin. Diabetes 2001; 50:
Inhibitory mechanism of vascular endothelial growth factor 1636–1642.
(VEGF) by bucillamine. Br J Pharmacol 2002; 137: 901–909. 167. Fang C, Jiang Z, Tomlinson DR. Expression of constitutive
150. Hikichi T, Mori F, Nakamura M, et al. Inhibitory effects of cyclo-oxygenase (COX-1) in rats with streptozotocin-induced
bucillamine on increased blood-retinal barrier permeability diabetes; effects of treatment with evening primrose oil
in streptozotocin-induced diabetic rats. Curr Eye Res 2002; 25: or an aldose reductase inhibitor on COX-1 mRNA levels.
1–7. Prostaglandins Leukot Essent Fatty Acids 1997; 56: 157–163.
151. Darnell JE. Transcription factors as targets for cancer therapy. 168. Giugliano D, Ceriello A, Paolisso G. Oxidative stress and
Nat Rev Cancer 2002; 2: 740–749. diabetic vascular complications. Diabetes Care 1996; 19:
152. Turkson J, Ryan D, Kim JS, et al. Phosphotyrosyl peptides block 257–267.
Stat3-mediated DNA binding activity, gene regulation, and cell 169. Kunisaki M, Bursell SE, Clermont AC, et al. Vitamin E prevents
transformation. J Biol Chem 2001; 276: 45 443–45 455. diabetes-induced abnormal retinal blood flow via the
153. Sowers JR. Effects of statins on the vasculature: implications for diacylglycerol-protein kinase C pathway. Am J Physiol 1995;
aggressive lipid management in the cardiovascular metabolic 269: E239–E246.
syndrome. Am J Cardiol 2003; 91: 14B–22B. 170. Bursell SE, Clermont AC, Aiello LP, et al. High-dose vitamin E
154. Liao JK. Beyond lipid lowering: the role of statins in vascular supplementation normalizes retinal blood flow and creatinine
protection. Int J Cardiol 2002; 86: 5–18. clearance in patients with type 1 diabetes. Diabetes Care 1999;
155. Delbosc S, Morena M, Djouad F, Ledoucen C, Descomps B, 22: 1245–1251.
Cristol JP. Statins, 3-hydroxy-3-methylglutaryl coenzyme A 171. Soriano FG, Pacher P, Mabley J, Liaudet L, Szabo C. Rapid
reductase inhibitors, are able to reduce superoxide anion reversal of the diabetic endothelial dysfunction by
production by NADPH oxidase in THP-1-derived monocytes. pharmacological inhibition of poly(ADP-ribose) polymerase.
J Cardiovasc Pharmacol 2002; 40: 611–617. Circ Res 2001; 89: 684–691.
156. Scalia R, Stalker TJ. Microcirculation as a target for the anti- 172. Kowluru RA, Koppolu P. Termination of experimental
inflammatory properties of statins. Microcirculation 2002; 9: galactosemia in rats, and progression of retinal metabolic
431–442. abnormalities. Invest Ophthalmol Vis Sci 2002; 43: 3287–3291.
157. Okamoto T, Yamagishi S, Inagaki Y, et al. Angiogenesis 173. Kowluru RA. Effect of reinstitution of good glycemic control
induced by advanced glycation end products and its prevention on retinal oxidative stress and nitrative stress in diabetic rats.
by cerivastatin. FASEB J 2002; 16: 1928–1930. Diabetes 2003; 52: 818–823.
158. Weis M, Heeschen C, Glassford AJ, Cooke JP. Statins have 174. Brooks SE, Gu X, Samuel S, et al. Reduced severity of oxygen-
biphasic effects on angiogenesis. Circulation 2002; 105: induced retinopathy in eNOS-deficient mice. Invest Ophthalmol
739–745. Vis Sci 2001; 42: 222–228.
159. Urbich C, Dernbach E, Zeiher AM, Dimmeler S. Double-edged 175. Ando A, Yang A, Mori K, et al. Nitric oxide is proangiogenic in
role of statins in angiogenesis signaling. Circ Res 2002; 90: the retina and choroid. J Cell Physiol 2002; 191: 116–124.
737–744.
160. Horiuchi M, Cui TX, Li Z, Li JM, Nakagami H, Iwai M.
Fluvastatin enhances the inhibitory effects of a selective

Copyright  2003 John Wiley & Sons, Ltd. Diabetes Metab Res Rev 2003; 19: 442–455.

You might also like