Europe PMC
Nothing Special   »   [go: up one dir, main page]

Europe PMC requires Javascript to function effectively.

Either your web browser doesn't support Javascript or it is currently turned off. In the latter case, please turn on Javascript support in your web browser and reload this page.

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


The central role of vascular endothelial growth factor (VEGF) signaling in regulating normal vascular development and pathological angiogenesis has been documented in multiple studies. Ocular anti-VEGF therapy is highly effective for treating a subset of patients with blinding eye disorders such as diabetic retinopathy and neovascular age-related macular degeneration (AMD). However, chronic VEGF suppression can lead to adverse effects associated with poor visual outcomes due to the loss of prosurvival and neurotrophic capacities of VEGF. In this review, we discuss emerging evidence for immune-related mechanisms that regulate ocular angiogenesis in a VEGF-independent manner. These novel molecular and cellular pathways may provide potential therapeutic avenues for a multitarget strategy, preserving the neuroprotective functions of VEGF in those patients whose disease is unresponsive to VEGF neutralization.

Free full text 


Logo of nihpaLink to Publisher's site
Trends Mol Med. Author manuscript; available in PMC 2016 Jan 1.
Published in final edited form as:
PMCID: PMC4282831
NIHMSID: NIHMS640418
PMID: 25457617

Seeing through VEGF: Innate and adaptive immunity in pathologic angiogenesis in the eye

Abstract

The central role of VEGF signaling in regulating normal vascular development and pathological angiogenesis has been documented in multiple studies. Ocular anti-VEGF therapy is highly effective for treating a subset of patients with blinding eye disorders such as diabetic retinopathy and neovascular age-related macular degeneration (AMD). However, chronic VEGF suppression can lead to adverse effects associated with poor visual outcomes due to the loss of pro-survival and neurotrophic capacities of VEGF. In this review, we discuss emerging evidence for immune-related mechanisms that regulate ocular angiogenesis in a VEGF-independent manner. These novel molecular and cellular pathways may provide potential therapeutic avenues for a multitarget strategy, preserving the neuroprotective functions of VEGF in those patients whose disease is unresponsive to VEGF neutralization.

Keywords: Angiogenesis, VEGF, immune regulation, neovascularization, AMD, diabetic retinopathy

Modulation of VEGF signaling: the therapeutic hub

Angiogenesis (see Glossary) is a physiological process involving the growth of new blood vessels from pre-existing vessels. In the eye, physiologic angiogenesis is critical during early development and indispensable for normal vision. In contrast, many disease states involving the eye such as diabetic retinopathy, retinal vein occlusions, age-related macular degeneration (AMD), and retinopathy of prematurity (ROP), have been linked to the disrupted regulation of this process leading to pathologic angiogenesis and blindness (Figure 1) [1].

An external file that holds a picture, illustration, etc.
Object name is nihms-640418-f0001.jpg
Anti-VEGF treatment of neovascular eye diseases

Fluorescein angiogram of a control eye (a), a patient with wet AMD (b) or proliferative diabetic retinopathy (PDR) (c). Bright hyperfluorescence demonstrates leakage of dye from the choroidal neovascularization (CNV) under the macula (b, arrow) and retinal neovascularization (c, arrows). Anomalous circulation characterized by neovascularization, ischemia and collateral vessel formation can be noted in the retina of PDR patient (c). Optical coherence tomography (OCT) of the central retina of an AMD patient before (d) and after (e) anti-VEGF treatments. Intraocular injections of anti-VEGF neutralizing antibody (e) led to resolution of cystoid macular edema (d, stars) and significant improvement in the CNV (d, arrowheads). OCT of neovascular AMD patient (f) shows a pigment epithelial detachment (PED) overlying the CNV (f, arrow). After intravitreal anti-VEGF treatment (g), there is an increase in the size of the PED (including the CNV complex) as well as the amount of subretinal fluid indicating persistent CNV activity (g, arrows).

Diabetes often results in significant tissue damage as manifested by nephropathy, neuropathy and in the eye, diabetic retinopathy. The systemic changes associated with elevated blood glucose and metabolic syndrome associated with diabetes lead to significant end organ damage in the eye, and especially in the retinal microcirculation [2]. Diabetic retinopathy is the leading cause of blindness in working age adults in the U.S., and it is associated with functional and structural alterations of retinal vasculature that later result in capillary degeneration and retinal ischemia. This is a public health crisis. In the U.S. alone, there are almost 750,000 people with proliferative diabetic retinopathy and over 60,000 new cases are reported each year. The earliest manifestation of diabetic retinopathy is in the form of pericyte loss that leads to instability of the retinal capillaries, formation of capillary microaneurysms, development of ischemic zones within the retina, and capillary leakage with subsequent retinal exudation and edema. Progressive damage to retinal microvascular network leads to tissue ischemia, upregulation of hypoxia inducible factor-1 (HIF-1), stimulation of vascular endothelial growth factor (VEGF) secretion and retinal neovascularization characterized by the development of new retinal and pre-retinal vascular fronds [3].

AMD is multifactorial disease and aging is the main risk factor. There are two forms of AMD; neovascular or wet AMD, and non-neovascular or dry AMD, which precedes the wet form [4]. Advanced AMD is characterized by an alteration of regulatory processes that either affect cell survival or induce cell proliferation, specifically neovascularization. AMD is the leading cause of blindness in people over 50 years of age in the industrialized world. Dry AMD is characterized by the development of lipid rich deposits called drusen underneath the retina and within the macula. The composition of these deposits has been reviewed recently [5]. As the disease progresses, drusen components induce inflammation and alterations in the retinal pigmented epithelium (RPE). In some patients, disease progresses to the advanced stages that can be associated with severe central vision loss. In advanced dry AMD, vision loss is secondary to RPE damage and cell death that subsequently leads to photoreceptor cell death and atrophic changes in the neurosensory retina. In the neovascular form, blindness is caused by pathogenic and proliferative neovascularization underneath the retina (choroidal neovascularization, CNV). The majority of acute vision loss in AMD is from the wet form of disease. These features can be overlapping as patients with advanced dry AMD can develop CNV and vice versa. In addition, as therapeutic agents targeted at factors that promote neovascularization become more efficacious, vision loss from atrophic retinal changes associated with dry AMD are becoming more prevalent [6].

Formerly known as retrolental fibroplasia, ROP was originally described in the 1940s by Terry, who first connected disease symptoms with premature birth [7]. At that time, no treatment for ROP was available. Major advances in ROP treatment became available in the 1980s and 1990s, when cryotherapy and laser photocoagulation of the avascular retina were shown to be partially effective in preventing blindness in ROP infants [8]. Although these treatments can reduce the incidence of blindness by 25%, patients are often left with poor visual acuity even after treatment. The pathogenesis of ROP has been associated with the use of high levels of oxygen on premature infants. The hyperoxia of the extra uterine environment, as well as the use of supplemented oxygen given to premature infants, slows or ceases retinal vascular growth that would normally occur in utero, and developed retinal vessels begin to regress [9]. As the infant matures, the non-vascularized retina becomes increasingly metabolically active, and in the absence of adequate vascularization, leads to tissue ischemia and hypoxia. This results in compensatory retinal neovascularization. New vessels form at the junction between the vascularized retina and the avascular zone of the retina. Over time, this pathological growth of vessels produces a fibrous scar extending from the retina to the vitreous gel and lens [9].

VEGF has been identified as one of the critical mediators of pathologic neovascularization. The role of VEGF in angiogenesis has been examined in great detail and is described in several review articles [10, 11]. In the eye, numerous studies have demonstrated the crucial role of VEGF during retinal vascular development [12, 13]. In this highly regulated process, VEGF is essential for the proliferation and migration of endothelial cells. It is also required for proper vascular patterning and tube formation (Figure 2). Additionally, the distinct vascular defects observed in experimental animal models depleted for alternative splice variants of VEGF confirmed the precise role of these isoforms in regulating retinal angiogenesis [14]. VEGF signaling has been attributed a key role in the maintenance of corneal transparency and the avascular state. Imbalance in VEGF related pathway is strongly associated with corneal neovascularization, a common clinical feature in diverse corneal diseases [15, 16]. More recent work demonstrated that VEGF also plays a prominent role in overall ocular development. Indeed, selective inactivation of VEGF expression in RPE resulted in a severe reduction of eyeball size associated with the loss of visual function [17]. RPE derived VEGF is also involved in the formation and maintenance of the choroidal vasculature that provides oxygen and nutrients to the outer retina [18].

An external file that holds a picture, illustration, etc.
Object name is nihms-640418-f0002.jpg
Role of VEGF in retinal angiogenesis and homeostasis

Schematic cross-section through the human eye (a) and detailed representation of the retina (b). In the retina, VEGF is produced and secreted by neurons, glia and vascular endothelial cells. Additionally, different cell types also express VEGF receptor. During retinal vascular development, VEGF signaling plays an essential role in the formation and patterning of the vascular network (vascular plexus). The retinal pigmented epithelium (RPE)-derived VEGF is responsible for the development and the maintenance of the choriocapillaries. It also provides neurotrophic support to photoreceptors, retinal bipolar cells (RBC) and retinal ganglion cells (RGC). Retinal VEGF expression is upregulated in response to hypoxia and contributes to pathologic neovascularization. In these conditions, the majority of the VEGF is produced by Müller glial cells and astrocytes.

The role of VEGF in ocular neovascular-related diseases has been characterized in experimental and translational studies [19]. Elevated intraocular VEGF is closely linked to the pathogenesis of several blinding eye diseases. In addition, early findings that the inhibition of VEGF signaling prevented neovascularization in ischemic retinal diseases highlighted the therapeutic relevance of anti-VEGF strategies in these conditions [20]. However, there is now substantial evidence showing that VEGF expression can also be induced through hypoxia-independent mechanisms [19].

Although VEGF clearly drives pathologic angiogenesis in disease, it also plays a crucial role in cellular homeostasis as a neurotrophic and cell survival factor [21-24]. Numerous cell types in the eye express and secrete VEGF, including endothelial cells, glial cells, retinal neurons, RPE, and immune cells such as monocytes/macrophages [25-28]. In disease states, intraocular VEGF expression is upregulated by hypoxia and inflammation [29]. VEGF-driven neovascularization and increased vascular permeability lead to complications including bleeding, retinal detachment and fibrovascular proliferation, all of which ultimately cause photoreceptor neuron death and blindness [1, 11]. As such, many treatment options for these complications are geared towards modifying VEGF levels within the eye in order to inhibit abnormal angiogenesis (Figure 1d,e). Numerous clinical trials have investigated therapies targeting VEGF and have shown efficacy in controlling disease and preventing vision loss (Table 1) [30-41].

Table1

Clincial trials targeting VEGFa

Study Disease Summary Anti-VEGF
Therapy
VISTA/VIVID Diabetes Randomized controlled trial (RCT) of
intravitreal aflibercept Injection for the
treatment of diabetic macular edema (DME)
[41]
Aflibercept: 97 kDa
recombinant fusion
protein consisting of
portions of human
VEGF receptors 1 and
2 extracellular domains
fused to the Fc portion
of human IgG1
COPERNICUS/
GALILEO
Vein occlusion RCT of intravitreal aflibercept injection in
patients with macular edema secondary
to retinal vein occlusions [37, 40]
RISE/RIDE Diabetes RCT of 0.3 or 0.5 mg intravitreal ranibizumab
for the treatment of DME [39]
BRAVO Vein occlusion RCT of 0.5 mg of intravitreal ranibizumab
injection in patients with macular edema
secondary to branch retinal vein occlusion
(BRVO) [34]
Ranibizumab: 48 kDa
recombinant
humanized IgG1 kappa
isotype monoclonal
antibody fragment that
binds to VEGF-A
CRUISE Vein occlusion RCT evaluating 0.5 mg of intravitreal
ranibizumab in patients with macular edema
[33] secondary to central retinal vein occlusion
(CRVO)
ANCHOR/
MARINA
AMD RCT of 0.3 or 0.5 mg of intravitreal
ranibizumab for neovascular AMD [31, 32]
Bevacizumab: 149 kDa
recombinant
humanized monoclonal
IgG1 antibody that
binds to VEGF-A
BEAT-ROP ROP RCT comparing intravitreal bevacizumab
monotherapy with conventional laser in
retinopathy of prematurity (ROP) [36]
VISION AMD RCT of intravitreal pegaptanib 0.3mg, 1.0mg
and 3.0mg for neovascular AMD [30]
Pegaptanib Na: a
pegylated aptamer that
targets the 165 isoform
of VEGF-A
CATT AMD A multicenter, noninferiority clinical trial
comparing 1.25 mg bevacizumab to 0.5 mg
ranibizumab in the treatment of neovascular
AMD [35]
-
VIEW1/2 AMD RCT comparing safety and efficacy of
intravitreal aflibercept to ranibizumab in
neovascular AMD [38]
-
aAbbreviations: AMD, age-related macular degeneration; BRVO, branch retinal vein occlusion; CRVO, central retinal vein occlusion; DME, diabetic macular edema; RCT, randomized controlled trial; ROP, retinopathy of prematurity; VEGF, vascular endothelial growth factor.

It is important to note that therapeutic VEGF inhibition leads to significant vision recovery in only a subset of patients with the retinal diseases highlighted above [42]. As such, VEGF-driven pathways are only a part of the complex machinery regulating angiogenesis in the eye [42]. This creates a large unmet need for developing novel strategies targeting pathways that regulate ocular angiogenesis that are independent of VEGF (Table 2). In addition, VEGF also plays a critical role in normal homeostasis in the eye in maintaining vascular integrity, as a neurotrophic and neuroprotective factor in the retina and the central nervous system, and indiscriminate or prolonged VEGF inhibition can lead to cell death and vision loss as demonstrated in animal models, cell culture and human randomized clinical trials [24, 42-44]. In this review, we will focus on VEGF-independent pathways involving the immune system that could provide additional insights into the regulation of normal and pathological angiogenesis in the eye.

Table2

Non-VEGF specific drugs that are being evaluated for inhibition of ocular angiogenesisa

Drug Disease Summary Mechanism
Pazopanib AMD 5 mg/mL Pazopanib eye drops resulted in mean
improvement in best-corrected visual acuity
(BCVA) at Day 29 and improvements in vision
[93]. Completed
Multitargeted tyrosine
kinase inhibitor of
VEGF receptors
(VEGFRs)-1, VEGFRs-
2, and VEGFRs-3,
platelet-derived growth
factor receptors
(PDGFRs)-α and
PDGFRs-β, and the
stem cell growth factor
c-Kit
Palomid 529 AMD Inhibits the TORC1 and TORC2 complexes and
shows both inhibition of Akt signaling and mTOR
signaling similarly in tumor and vasculature.
Shows that P529 inhibits tumor growth,
angiogenesis, and vascular permeability [94].
Completed
Dual TORC1/2 inhibitor
of the P13K/Akt/m TOR
pathway
Squalamine
Lactate
AMD Systemically administered squalamine lactate
partially reduced choroidal neovascular
membrane development induced by laser trauma
in experimental model [95]. Ongoing
An inhibitor of new
blood vessel formation
(angiogenesis) induced
by VEGF, PDGF or
bFGF.
AdGVPEDF.11D AMD Adenoviral vectored PEDF (AdGVPEDF.11D)
administered bysubtenon injection,
inhibits choroidal neovascularization in experimental
model [96]. Completed.
adenovirus vector
containing the gene for
the PEDF (pigment
epithelium-derived
factor) protein
Anecortave
acetate
AMD Anecortave acetate 15 mg has been shown to be
efficacious at inhibiting vision loss and choroidal
neovascularization lesion growth in a placebo-
controlled, double-masked clinical trial in patients
with exudative age-related macular degeneration
[97]. Completed
Anecortave acetate
blocks proteolytic
cascade in the vascular
endothelial cells
Fovista AMD Fovista has been administered in combination
with an anti-VEGF agent (ranibizumab) for the
treatment of patients newly diagnosed with wet
AMD [98]. Recruiting
An anti-PDGF aptamer
that prevent the binding
of PDGF to its
receptors on pericytes
Palomid 529 AMD Subconjontival injection in neovascular AMD
patients that have not responded to standard anti-
VEGF treatments [99]. Completed
An inhibitor of mTOR
production
OC-10X Diabetes Evaluate the safety and tolerability of topical OC-
10X Ophthalmic Suspension in healthy human
subjects. OC-10X is under development for the
treatment of Proliferative Diabetic Retinopathy
(PDR). Completed
Oc-10X is a selective
tubulin inhibitor
TG100801 AMD/
Diabetes
Topical treatment for inhibition of ocular
angiogenesis and retinal edema [100]. Completed
Inhibitor of Src kinases
and selected receptor
tyrosine kinases
aAbbreviations: AMD, age-related macular degeneration; BCVA, best-corrected visual acuity; bFGF, basic fibroblast growth factor; PEDF, pigment epithelium-derived factor; PDGF, platelet-derived growth factor; PDR, proliferative diabetic retinopathy; VEGF, vascular endothelial growth factor.

Innate immunity and ocular angiogenesis

In both physiological and pathological conditions, the innate immune system has been shown to play a determinant role in the vascular remodeling process. The link between inflammation and angiogenesis has been defined in the pathophysiology of several diseases, including cancer, atherosclerosis and eye disorders [1, 5, 45]. In these conditions, activation of innate immunity is often an early event in the disease process that further guides the local tissue microenvironment and ultimately molds cellular and molecular responses. The crucial role of VEGF signaling in ocular angiogenesis has been extensively studied and the molecular response of endothelial cells to VEGF is well documented [1, 10, 11]. There is also a wealth of evidence implicating innate immunity in key processes during angiogenesis, including endothelial cell proliferation, migration and vessel anastomosis [46]. The concomitant activation of an innate immune response, associated with dysregulated blood vessel growth (as observed in many eye diseases), and the implication of inflammatory mediators in VEGF signaling have led to the belief that VEGF plays an omnipotent role in regulating ocular angiogenesis. Although recent translational studies targeting VEGF have confirmed its pivotal role in blinding eye diseases, there is emerging evidence highlighting VEGF-independent pathways in disease pathogenesis that suggest potentially novel approaches involving components of innate immunity that may modulate pathological angiogenesis in the eye.

Convergent complement pathways

The complement system is composed of three distinct pathways (classical, lectin, and alternative), the activation of which contributes to many aspects of immune responses. Defective complement components are causative in various pathologies, extending far beyond the autoimmune diseases [47]. For instance, polymorphisms in numerous complement pathway genes are strongly associated with neurodegenerative disorders such as AMD [48]. Recent studies have demonstrated that complement pathways tightly regulate pathological neovascularization in the retina, suggesting interplay between innate immunity and ocular angiogenic processes [49, 50].

Oxygen-induced retinopathy (OIR; Box 1) is widely used in animal models of pathological vascularization to mimic clinical features of proliferative retinopathies, including those observed in ROP, a leading cause of vision loss in children [51]. Specific targeting of C3 and C5, key components of the complement response, directly implicated this pathway in the biology of normal and pathological retinal blood vessel growth [49]. In the context of OIR, C3 deficiency induced an increase of pathological angiogenesis in the retina. In addition, selective depletion or functional blockade of the active component C5a also resulted in higher neovascularization, suggesting that activation of the complement system prevented retinal neovascularization [49]. Although the anti-angiogenic effects of C3 and C5 are mediated by macrophages, the effective pathways regulating retinal angiogenesis are slightly different. Macrophage depletion in vivo abolished the complement-dependent repression of neovascularization. Furthermore, complement activation polarized macrophages to an anti-angiogenic phenotype that directly inhibited endothelial cell proliferation [49].

Box1

Experimental models of ocular angiogenesis

Oxygen induced retinopathy (OIR)

Ischemia induced neovascularization is a common feature of many retinal disorders including retinopathy of prematurity (ROP) and diabetic retinopathy (DR). In ROP, high oxygen exposure results in vaso-obliteration of central retinal vessels. When returned to normal oxygen conditions, hypoxic retinal areas develop new blood vessels [9]. This two-step neovascularization is also observed in patients with DR where insulin resistance and other metabolic factors cause hyperglycemia that induces vascular occlusion and the subsequent retinal ischemia promotes neovessel formation [90]. Multiple experimental procedures now recapitulate the sequential events that lead to pathological angiogenesis in animal models. Neonatal mice are exposed to high oxygen (75%) from postnatal days (P) 7 to 12, corresponding to the vaso-obliteration phase, and are then placed in normal room air (21% oxygen) for 5 additional days, where vessel proliferation occurs. At P17, there is spontaneous neovessel regression [91]. OIR is intensively used to study retinal neovascularization in a pathological context and to characterize mechanisms modulating blood vessel homeostasis [51].

Injury-induced choroidal neovascularization (CNV)

There are two forms of age related macular degeneration (AMD), a major cause of blindness in the elderly [4]. In advanced AMD the dry or non-neovascular form is characterized by retinal pigmented epithelial cell death and subsequent photoreceptor neuron loss associated with atrophic changes in the macula, whereas in the wet or neovascular form, there is a formation of new blood vessels underneath the retina called choroidal neovascularization (CNV). Although the dry form is more common, the majority of blindness occurs in patients with the neovascular form of AMD, due to macular edema and exudation of fluid and blood from CNV. To date, there is no animal model that recapitulates all the clinical features of AMD. In experimental models, to mimic CNV formation as seen in the wet form of AMD, neovessels are induced in the subretinal space after focal laser-induced disruption of the Bruch’s membrane. A week after laser-induced injury, neovascularization is analyzed by immunohistochemistry [92].

The findings that complement inhibits the formation of pathological neovessels have been confirmed by recent studies specifically targeting the alternative pathway. Mice lacking the complement factor B (cfB) developed more neovessels in the OIR model [50]. In addition, deletion of cfB had no effect on VEGF signaling, suggesting that this complement component could directly target endothelial cells [50]. Early studies have demonstrated that cfB interaction with the membrane bound C3b is a key step that initiates the formation of the C3 convertase complex leading to death of the target cell after activation of the membrane attack complex [52, 53]. Interestingly, cfB was associated with retinal neovessels, whereas the convertase inhibitor (CD55 or decay-accelerating factor) was restricted to the normal vasculature [50]. Indeed, cfB deficiency altered the targeting of pathological endothelial cells by the complement alternative pathway, and resulted in an accumulation of neovessels. Given the involvement of complement system in diverse biological processes, the selective activation of a complement pathway, combined with specific cell targeting, provides a regulatory mechanism that prevents collateral tissue damage and harm to the host organism [50].

Macrophages: from effector to mediator

It is now well established that macrophages play a significant role in regulating ocular angiogenesis through multiple mechanisms [54-59]. As shown above, macrophages can also mediate the effect of activated complement system on retinal vasculature. Depending on the tissue context, macrophages can be classically (M1) or alternatively (M2) activated. These macrophage subpopulations are characterized by a variety of specific markers and exhibit differential cytokine production, receptor expression, and effector function [60-64]. A more recent macrophage classification has been proposed based on their activation conditions [64]. Pro-inflammatory M1 macrophages are anti-angiogenic, and express high levels of tumor necrosis factor alpha (TNF-α), interleukin-12 (IL-12), IL-6, IL-1β, inducible nitric oxide synthase (iNOS) and matrix metalloproteinase-9 (MMP9). In contrast, pro-angiogenic M2 macrophages mediate wound healing, and are characterized by low M1 signature markers but increased expression of IL-10, CD163, and transforming growth factor beta (TGF-β). In an animal model of neovascular AMD, it has been demonstrated that the switch of macrophage polarization from M1 to M2 is responsible for the increase of choroidal neovascularization (CNV) in old mice [65]. A similar dynamic and functional switch was also observed in AMD patients with alternatively activated ocular macrophages in advanced stages of the disease [66, 67].

In early studies, it was suggested that macrophages inhibited CNV through Fas ligand (CD95) signaling. However, these findings did not rule out a possible contribution of VEGF pathways in this pathological angiogenesis [54, 68]. Subsequently it has been demonstrated that in addition to aging [65], dysregulation of cholesterol metabolism polarized macrophages to a pro-angiogenic phenotype [69]. Further characterization of mice lacking a key regulator of cholesterol homeostasis confirmed that impaired macrophage regulation of endothelial cells growth was VEGF-independent [69].

In the retina, resident macrophages (microglial cells) are essential for proper blood vessel formation [55]. During development or in a pathological context, remodeling of the retinal vasculature by microglial cells is mediated by secreted angiogenic factors, cytokines or regulators of the extracellular matrix (ECM). Recent identification of VEGF-independent angiogenesis through neuropilin-1 (NRP1) signaling provided new insights into the interplay between endothelial and surrounding retinal cells [70]. Activation of NRP1 by ECM signals induced a phosphorylation of integrin targets such as paxillin and subsequent actin remodeling which promoted endothelial cell motility [70]. Although NRP1 is a VEGF co-receptor, this study clearly demonstrated that selective targeting of ECM–stimulated NRP1 signaling altered the regulation of actin cytoskeleton and cell migration. These findings suggested that NRP1 signaling is involved in both physiological and pathological angiogenesis in the eye [70]. Similarly, previous studies showed that macrophage modulation of ECM proteins is critical for the retinal vascular remodeling [57]. During pathological conditions, macrophage metalloproteinases (MMP-2 and 9) promoted retinal neovascularization by directly targeting the ECM [57].

Despite the close interaction between macrophages and retinal blood vessels, the effect of macrophages on endothelial cells can be modulated through indirect mechanisms. Findings from a recent study showed that microglial cells regulated neovessel growth through inflammatory mediators that target adjacent neurons [71]. In the OIR model, activation of microglial cells resulted in overexpression of IL-1β, which in turn induced Semaphorin-3a (Sema-3A) production by retinal ganglion cells through IL-1R signaling. The secreted pro-apoptotic factor Sema-3A then induced endothelial cell death. Furthermore, selective blockade of IL-1β signaling prevents this cytotoxic cascade that leads to vascular degeneration [71]. The above findings demonstrate that although microglial cells contribute to retinal vascular development, they also play a pivotal role in modulating retinal neovascularization.

The role of adaptive immunity in ocular angiogenesis

Recent studies have suggested that the adaptive immune compartment may play a role in promoting pathologic angiogenesis in the eye [72-75]. A number of studies have suggested that Th17 cells play a role in advanced AMD [76-78]. Th17 cells are effector CD4+ T cells that preferentially produce IL-17 upon specific surface markers stimulation or activation by various pathogens. IL-17 is a pro-inflammatory cytokine that mostly targets epithelial, endothelial and stromal cells and defective production of this cytokine is associated with several autoimmune diseases [79, 80]. Complement components such as C5a have been implicated in inducing CD4+ T cells to express IL-17 and IL-22. The level of these cytokines was also increased in serum from patients with AMD [76]. In addition, one study showed that the IL-17RC promoter, a component of the IL-17R complex, was significantly hypomethylated in patients with AMD [81]. This was associated with increased expression of IL-17RC in the peripheral blood, retina and choroid of AMD patients [81]. A subsequent study that carefully examined the methylation status of the IL-17RC promoter using global methylation arrays, bisulfite sequencing and PCR-based methylation assays found no significant difference between AMD patients and age-matched controls in two independent cohorts [82]. These findings were consistent for no difference either in the peripheral blood or in eye-derived tissues such as the retina or choroid [82]. Furthermore, it was demonstrated that IL-17 alone had no impact on vessel growth, although it can potentiate the effect of other angiogenic factors such as VEGF, basic fibroblast growth factor (bFGF) and hepatocyte growth factor (HGF) [72]. Recent findings have shown that combined production of IL-17 from γδT cells and innate lymphoid cells was responsible for the increased neovascularization observed in experimental model of CNV [74]. As such, the precise role of IL-17 in regulating angiogenesis in the eye remains unclear.

Complement components and lipid peroxidation can induce oxidation specific epitopes that may function as antigens for immune recognition [83]. Carboxyethyl pyrrole (CEP) is an example of such an epitope that is an oxidation fragment of docohexaenoic acid (DHA) [84]. CEP has previously been shown in animals to initiate an immune response that leads to damage to the RPE similar to that seen in non-neovascular AMD [85-87]. In addition, not only is CEP increased in AMD donor eyes, but autoantibodies to CEP and CEP-modified proteins can be identified in plasma from AMD patients [88, 89]. The mechanism by which CEP induces an immune response is thought to be via the activation of T lymphocytes by classically activated M1 macrophages [77]. In murine models, this leads to increased expression of IFN-γ and IL-17 [77]. Despite the association of CEP and similar epitopes with the disease phenotype in dry (non-neovascular) AMD, the link to pathologic angiogenesis and wet AMD remains elusive.

Autoantibody profiling of sera from AMD patients has identified retinal autoantibodies that have the ability to promote endothelial cell growth in vitro. Although underlying mechanisms that lead to endothelial cell proliferation are unknown, these findings suggest that autoantibodies could be involved in the pathogenesis of neovascular AMD and especially in CNV formation [73].

Concluding remarks and future perspectives

It is now apparent that chronic, intraocular anti-VEGF treatments could lead to permanent vision loss due to the pleiotropic effects of VEGF and, more importantly, its neurotrophic potential. Both experimental and translational studies have confirmed the deleterious effects of long-term VEGF inhibition. There are several lines of evidence for VEGF-independent regulation of ocular angiogenesis. Different components of the immune system have been shown to modulate vascularization in the eye. Although some regulatory pathways are specific to the innate or adaptive compartment, the interplay between immune components and retinal cells is necessary for the control of ocular angiogenesis (Figure 3). For instance, macrophages/microglia can directly regulate endothelial cells growth upon activation, and also mediate the effects of the complement system. The impact of immune responses on retinal vasculature can be facilitated or enhanced by surrounding neurons. The latter shows the interconnection between the immune system, retinal vasculature and neurons. Future studies will determine the impact of selective inhibition of ocular angiogenesis on neuron survival and local immune responses (Box 2).

Box2

Outstanding questions

  • What is the role of local neurons in mediating angiogenesis in the eye?

  • How does immune privilege affect the regulation of ocular angiogenesis?

  • Does blood retinal barrier integrity correlate with development of retinal autoantibodies?

  • Is aging of the immune system causative in vascular proliferative diseases?

An external file that holds a picture, illustration, etc.
Object name is nihms-640418-f0003.jpg
Immune regulation of VEGF-independent angiogenesis

Depending on their activation state, macrophages/microglia can differentially modulate endothelial cell growth. M1 macrophages are anti-angiogenic, whereas M2 macrophages promote angiogenesis. The effect of macrophages on endothelial cells can be contact-dependent or through secretion of angiogenic factors. Complement factor B (cfB), a component of the alternative complement pathway, in combination with the complement inhibitor (CD55), selectively inhibits pathological neovessel formation. In hypoxic conditions, CD55 is downregulated in endothelial cells therefore allowing the specific targeting of neovessels by cfB. Complement components can also modulate macrophage activation. Complement components C3 and C5 polarize macrophages to an M1 anti-angiogenic phenotype. Activation of microglial cells upon exposure to ischemia induces the secretion of IL-1β and subsequent production of Semaphorin-3a (Sema-3A) by retinal neurons. Sema-3A induces degeneration of the retinal vasculature. The adaptive immunity may promote angiogenesis through autoantibodies or secretion of IL-17 by T cells. Ocular angiogenesis can be further regulated through the remodeling of the extracellular matrix (ECM). Activation of ECM-stimulated pathways through neuropilin-1 (NRP1) signaling also promotes angiogenesis in the eye.

  • VEGF in eye diseases

  • Immune regulation of pathologic neovascularization

  • VEGF-independent therapies in retinal vascular diseases

Acknowledgments

This work was supported by NIH grant R01EY019287; NIH Vision Core Grant P30EY02687; the Carl Marshall Reeves and Mildred Almen Reeves Foundation Inc. Award; the Research to Prevent Blindness Inc. Career Development Award and Physician Scientist Award to Dr. Apte; the American Federation for Aging Research; the Lacy Foundation Research Award and the Thome Foundation.

Glossary

Angiogenesisbiological process of blood vessel formation
Adaptive immunityspecific antibodies or cell-mediated responses are developed after exposure to pathogens/antigens. This adaptive line of defense also produces an immunological memory
Complementthe complement system is a component of the innate immunity and comprises a network of proteins and receptors that recognizes pathogens
Choroidal neovascularization (CNV)Pathological angiogenesis that develops underneath the retina. These new vessels are responsible for the hemorrhage and leakage of fluid causing photoreceptor death, neuroretinal degeneration and blindness
Immune systemis a network of specialized cells and structures that orchestrate the defense of the body against infections and diseases. It has innate and adaptive components
InflammationA cascade of events at the site of infection or tissue injury that lead to the recruitment of immune cells and secretion of soluble factors including cytokines and chemokines for the resolution and limitation of tissue damage
Innate immunitythis defense mechanism is not specific to pathogens, and recognizes common features of invading organisms
Ischemiadeficient blood supply to tissues. In vascular proliferative eye diseases such as retinopathy of prematurity and diabetic retinopathy, ischemia is a key signal that triggers neovascularization
Retinais the light sensitive, transparent tissue lining the back of the eye. It is composed of different cells (neurons and glia) organized in well-structured layers. The photoreceptor neurons transform light photons into electrical signals for further processing in the brain
Vascular endothelial growth factor (VEGF)an important regulator of developmental and pathological angiogenesis.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

1. Rajappa M, et al. Ocular angiogenesis: mechanisms and recent advances in therapy. Advances in clinical chemistry. 2010;50:103–121. [Abstract] [Google Scholar]
2. Safi SZ, et al. Molecular Mechanisms of Diabetic Retinopathy, General Preventive Strategies, and Novel Therapeutic Targets. BioMed research international. 2014;2014:801269. [Europe PMC free article] [Abstract] [Google Scholar]
3. Chou J, et al. Role of endothelial cell and pericyte dysfunction in diabetic retinopathy: review of techniques in rodent models. Advances in experimental medicine and biology. 2014;801:669–675. [Europe PMC free article] [Abstract] [Google Scholar]
4. Friedman DS, et al. Prevalence of age-related macular degeneration in the United States. Archives of ophthalmology. 2004;122:564–572. [Abstract] [Google Scholar]
5. Sene A, Apte RS. Eyeballing cholesterol efflux and macrophage function in disease pathogenesis. Trends in endocrinology and metabolism: TEM. 2014;25:107–114. [Europe PMC free article] [Abstract] [Google Scholar]
6. Klein R, et al. The epidemiology of age-related macular degeneration. American journal of ophthalmology. 2004;137:486–495. [Abstract] [Google Scholar]
7. Terry TL. Fibroblastic Overgrowth of Persistent Tunica Vasculosa Lentis in Infants Born Prematurely: II. Report of Cases-Clinical Aspects. Transactions of the American Ophthalmological Society. 1942;40:262–284. [Europe PMC free article] [Abstract] [Google Scholar]
8. Multicenter trial of cryotherapy for retinopathy of prematurity. Preliminary results. Cryotherapy for Retinopathy of Prematurity Cooperative Group. Archives of ophthalmology. 1988;106:471–479. [Abstract] [Google Scholar]
9. Mechoulam H, Pierce EA. Retinopathy of prematurity: molecular pathology and therapeutic strategies. American journal of pharmacogenomics : genomics-related research in drug development and clinical practice. 2003;3:261–277. [Abstract] [Google Scholar]
10. Olsson AK, et al. VEGF receptor signalling - in control of vascular function. Nature reviews. Molecular cell biology. 2006;7:359–371. [Abstract] [Google Scholar]
11. Qazi Y, et al. Mediators of ocular angiogenesis. Journal of genetics. 2009;88:495–515. [Europe PMC free article] [Abstract] [Google Scholar]
12. Dorrell MI, et al. Retinal vascular development is mediated by endothelial filopodia, a preexisting astrocytic template and specific R-cadherin adhesion. Investigative ophthalmology & visual science. 2002;43:3500–3510. [Abstract] [Google Scholar]
13. Dorrell MI, Friedlander M. Mechanisms of endothelial cell guidance and vascular patterning in the developing mouse retina. Progress in retinal and eye research. 2006;25:277–295. [Abstract] [Google Scholar]
14. Stalmans I, et al. Arteriolar and venular patterning in retinas of mice selectively expressing VEGF isoforms. The Journal of clinical investigation. 2002;109:327–336. [Europe PMC free article] [Abstract] [Google Scholar]
15. Bock F, et al. Novel anti(lymph)angiogenic treatment strategies for corneal and ocular surface diseases. Progress in retinal and eye research. 2013;34:89–124. [Abstract] [Google Scholar]
16. Cursiefen C, et al. Consensus statement on indications for anti-angiogenic therapy in the management of corneal diseases associated with neovascularisation: outcome of an expert roundtable. The British journal of ophthalmology. 2012;96:3–9. [Abstract] [Google Scholar]
17. Marneros AG, et al. Vascular endothelial growth factor expression in the retinal pigment epithelium is essential for choriocapillaris development and visual function. American Journal of Pathology. 2005;167:1451–1459. [Europe PMC free article] [Abstract] [Google Scholar]
18. Saint-Geniez M, et al. VEGF expression and receptor activation in the choroid during development and in the adult. Investigative ophthalmology & visual science. 2006;47:3135–3142. [Abstract] [Google Scholar]
19. Penn JS, et al. Vascular endothelial growth factor in eye disease. Progress in retinal and eye research. 2008;27:331–371. [Europe PMC free article] [Abstract] [Google Scholar]
20. Aiello LP, et al. Suppression of retinal neovascularization in vivo by inhibition of vascular endothelial growth factor (VEGF) using soluble VEGF-receptor chimeric proteins. Proceedings of the National Academy of Sciences of the United States of America. 1995;92:10457–10461. [Europe PMC free article] [Abstract] [Google Scholar]
21. Robinson GS, et al. Nonvascular role for VEGF: VEGFR-1, 2 activity is critical for neural retinal development. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2001;15:1215–1217. [Abstract] [Google Scholar]
22. Jin K, et al. Vascular endothelial growth factor (VEGF) stimulates neurogenesis in vitro and in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:11946–11950. [Europe PMC free article] [Abstract] [Google Scholar]
23. Zachary I. Neuroprotective role of vascular endothelial growth factor: signalling mechanisms, biological function, and therapeutic potential. Neuro-Signals. 2005;14:207–221. [Abstract] [Google Scholar]
24. Kurihara T, et al. Targeted deletion of Vegfa in adult mice induces vision loss. The Journal of clinical investigation. 2012;122:4213–4217. [Europe PMC free article] [Abstract] [Google Scholar]
25. Kim I, et al. Constitutive expression of VEGF, VEGFR-1, and VEGFR-2 in normal eyes. Investigative ophthalmology & visual science. 1999;40:2115–2121. [Abstract] [Google Scholar]
26. Tsutsumi C, et al. The critical role of ocular-infiltrating macrophages in the development of choroidal neovascularization. Journal of leukocyte biology. 2003;74:25–32. [Abstract] [Google Scholar]
27. Marneros AG, et al. Vascular endothelial growth factor expression in the retinal pigment epithelium is essential for choriocapillaris development and visual function. The American journal of pathology. 2005;167:1451–1459. [Europe PMC free article] [Abstract] [Google Scholar]
28. Sapieha P. Eyeing central neurons in vascular growth and reparative angiogenesis. Blood. 2012;120:2182–2194. [Abstract] [Google Scholar]
29. Vadlapatla RK, et al. Hypoxia-inducible factor-1 (HIF-1): a potential target for intervention in ocular neovascular diseases. Current drug targets. 2013;14:919–935. [Europe PMC free article] [Abstract] [Google Scholar]
30. Gragoudas ES, et al. Pegaptanib for neovascular age-related macular degeneration. New Engl J Med. 2004;351:2805–2816. [Abstract] [Google Scholar]
31. Brown DM, et al. Ranibizumab versus verteporfin for neovascular age-related macular degeneration. New Engl J Med. 2006;355:1432–1444. [Abstract] [Google Scholar]
32. Rosenfeld PJ, et al. Ranibizumab for neovascular age-related macular degeneration. New Engl J Med. 2006;355:1419–1431. [Abstract] [Google Scholar]
33. Brown DM, et al. Ranibizumab for macular edema following central retinal vein occlusion: six-month primary end point results of a phase III study. Ophthalmology. 2010;117:1124–1133. e1121. [Abstract] [Google Scholar]
34. Campochiaro PA, et al. Ranibizumab for Macular Edema following Branch Retinal Vein Occlusion Six-Month Primary End Point Results of a Phase III Study. Ophthalmology. 2010;117:1102–U1111. [Abstract] [Google Scholar]
35. Martin DF, et al. Ranibizumab and Bevacizumab for Neovascular Age-Related Macular Degeneration The CATT Research Group. New Engl J Med. 2011;364:1897–1908. [Europe PMC free article] [Abstract] [Google Scholar]
36. Mintz-Hittner HA, et al. Efficacy of Intravitreal Bevacizumab for Stage 3+Retinopathy of Prematurity. New Engl J Med. 2011;364:603–615. [Europe PMC free article] [Abstract] [Google Scholar]
37. Boyer D, et al. Vascular Endothelial Growth Factor Trap-Eye for Macular Edema Secondary to Central Retinal Vein Occlusion Six-Month Results of the Phase 3 COPERNICUS Study. Ophthalmology. 2012;119:1024–1032. [Abstract] [Google Scholar]
38. Heier JS, et al. Intravitreal Aflibercept (VEGF Trap-Eye) in Wet Age-related Macular Degeneration. Ophthalmology. 2012;119:2537–2548. [Abstract] [Google Scholar]
39. Brown DM, et al. Long-term Outcomes of Ranibizumab Therapy for Diabetic Macular Edema: The 36-Month Results from Two Phase III Trials RISE and RIDE. Ophthalmology. 2013;120:2013–2022. [Abstract] [Google Scholar]
40. Holz FG, et al. VEGF Trap-Eye for macular oedema secondary to central retinal vein occlusion: 6-month results of the phase III GALILEO study. Brit J Ophthalmol. 2013;97:278–284. [Abstract] [Google Scholar]
41. Midena E, Invest VDVDS. Intravitreal Aflibercept for the Treatment of Diabetic Macular Edema (Dme): Findings from the Vivid-Dme and Vista-Dme Clinical Trials. Eur J Ophthalmol. 2014;24:459–459. [Google Scholar]
42. Rofagha S, et al. Seven-year outcomes in ranibizumab-treated patients in ANCHOR, MARINA, and HORIZON: a multicenter cohort study (SEVEN-UP) Ophthalmology. 2013;120:2292–2299. [Abstract] [Google Scholar]
43. Pelletier J, et al. VEGF-A Promotes Both Pro-angiogenic and Neurotrophic Capacities for Nerve Recovery After Compressive Neuropathy in Rats. Molecular neurobiology. 2014 [Abstract] [Google Scholar]
44. Park HY, et al. Neuronal cell death in the inner retina and the influence of vascular endothelial growth factor inhibition in a diabetic rat model. The American journal of pathology. 2014;184:1752–1762. [Abstract] [Google Scholar]
45. Newman AC, Hughes CC. Macrophages and angiogenesis: a role for Wnt signaling. Vascular cell. 2012;4:13. [Europe PMC free article] [Abstract] [Google Scholar]
46. Frantz S, et al. Innate immunity and angiogenesis. Circulation research. 2005;96:15–26. [Abstract] [Google Scholar]
47. Ricklin D, et al. Complement: a key system for immune surveillance and homeostasis. Nature immunology. 2010;11:785–797. [Europe PMC free article] [Abstract] [Google Scholar]
48. Parmeggiani F, et al. Mechanism of inflammation in age-related macular degeneration: an up-to-date on genetic landmarks. Mediators Inflamm. 2013;2013:435607. [Europe PMC free article] [Abstract] [Google Scholar]
49. Langer HF, et al. Complement-mediated inhibition of neovascularization reveals a point of convergence between innate immunity and angiogenesis. Blood. 2010;116:4395–4403. [Europe PMC free article] [Abstract] [Google Scholar]
50. Sweigard JH, et al. The alternative complement pathway regulates pathological angiogenesis in the retina. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2014;28:3171–3182. [Europe PMC free article] [Abstract] [Google Scholar]
51. Stahl A, et al. The Mouse Retina as an Angiogenesis Model. Investigative ophthalmology & visual science. 2010;51:2813–2826. [Europe PMC free article] [Abstract] [Google Scholar]
52. Muller-Eberhard HJ. The membrane attack complex of complement. Annual review of immunology. 1986;4:503–528. [Abstract] [Google Scholar]
53. Forneris F, et al. Structures of C3b in complex with factors B and D give insight into complement convertase formation. Science. 2010;330:1816–1820. [Europe PMC free article] [Abstract] [Google Scholar]
54. Apte RS, et al. Macrophages inhibit neovascularization in a murine model of age-related macular degeneration. PLoS medicine. 2006;3:e310. [Abstract] [Google Scholar]
55. Checchin D, et al. Potential role of microglia in retinal blood vessel formation. Investigative ophthalmology & visual science. 2006;47:3595–3602. [Abstract] [Google Scholar]
56. Fantin A, et al. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF-mediated endothelial tip cell induction. Blood. 2010;116:829–840. [Europe PMC free article] [Abstract] [Google Scholar]
57. Kubota Y, et al. M-CSF inhibition selectively targets pathological angiogenesis and lymphangiogenesis. The Journal of experimental medicine. 2009;206:1089–1102. [Europe PMC free article] [Abstract] [Google Scholar]
58. Lobov IB, et al. WNT7b mediates macrophage-induced programmed cell death in patterning of the vasculature. Nature. 2005;437:417–421. [Europe PMC free article] [Abstract] [Google Scholar]
59. Outtz HH, et al. Notch1 controls macrophage recruitment and Notch signaling is activated at sites of endothelial cell anastomosis during retinal angiogenesis in mice. Blood. 2011;118:3436–3439. [Europe PMC free article] [Abstract] [Google Scholar]
60. Mosser DM. The many faces of macrophage activation. Journal of leukocyte biology. 2003;73:209–212. [Abstract] [Google Scholar]
61. Mantovani A, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends in immunology. 2004;25:677–686. [Abstract] [Google Scholar]
62. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. The Journal of clinical investigation. 2012;122:787–795. [Europe PMC free article] [Abstract] [Google Scholar]
63. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000prime reports. 2014;6:13. [Europe PMC free article] [Abstract] [Google Scholar]
64. Murray PJ, et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41:14–20. [Europe PMC free article] [Abstract] [Google Scholar]
65. Kelly J, et al. Senescence regulates macrophage activation and angiogenic fate at sites of tissue injury in mice. The Journal of clinical investigation. 2007;117:3421–3426. [Europe PMC free article] [Abstract] [Google Scholar]
66. Cherepanoff S, et al. Bruch's membrane and choroidal macrophages in early and advanced age-related macular degeneration. The British journal of ophthalmology. 2010;94:918–925. [Abstract] [Google Scholar]
67. Cao X, et al. Macrophage polarization in the maculae of age-related macular degeneration: a pilot study. Pathology international. 2011;61:528–535. [Europe PMC free article] [Abstract] [Google Scholar]
68. Roychoudhury J, et al. Targeting immune privilege to prevent pathogenic neovascularization. Investigative ophthalmology & visual science. 2010;51:3560–3566. [Europe PMC free article] [Abstract] [Google Scholar]
69. Sene A, et al. Impaired cholesterol efflux in senescent macrophages promotes age-related macular degeneration. Cell metabolism. 2013;17:549–561. [Europe PMC free article] [Abstract] [Google Scholar]
70. Raimondi C, et al. Imatinib inhibits VEGF-independent angiogenesis by targeting neuropilin 1-dependent ABL1 activation in endothelial cells. The Journal of experimental medicine. 2014;211:1167–1183. [Europe PMC free article] [Abstract] [Google Scholar]
71. Rivera JC, et al. Microglia and interleukin-1beta in ischemic retinopathy elicit microvascular degeneration through neuronal semaphorin-3A. Arteriosclerosis, thrombosis, and vascular biology. 2013;33:1881–1891. [Abstract] [Google Scholar]
72. Takahashi H, et al. Interleukin-17 enhances bFGF-, HGF- and VEGF-induced growth of vascular endothelial cells. Immunology letters. 2005;98:189–193. [Abstract] [Google Scholar]
73. Morohoshi K, et al. Serum autoantibody biomarkers for age-related macular degeneration and possible regulators of neovascularization. Experimental and molecular pathology. 2012;92:64–73. [Abstract] [Google Scholar]
74. Hasegawa E, et al. IL-23-independent induction of IL-17 from gammadeltaT cells and innate lymphoid cells promotes experimental intraocular neovascularization. Journal of immunology. 2013;190:1778–1787. [Abstract] [Google Scholar]
75. Camelo S. Potential Sources and Roles of Adaptive Immunity in Age-Related Macular Degeneration: Shall We Rename AMD into Autoimmune Macular Disease? Autoimmune diseases. 2014;2014:532487. [Europe PMC free article] [Abstract] [Google Scholar]
76. Liu B, et al. Complement component C5a promotes expression of IL-22 and IL-17 from human T cells and its implication in age-related macular degeneration. Journal of translational medicine. 2011;9:1–12. [Europe PMC free article] [Abstract] [Google Scholar]
77. Cruz-Guilloty F, et al. T cells and macrophages responding to oxidative damage cooperate in pathogenesis of a mouse model of age-related macular degeneration. PloS one. 2014;9:e88201. [Europe PMC free article] [Abstract] [Google Scholar]
78. Nussenblatt RB, et al. Immune Responses in Age-Related Macular Degeneration and a Possible Long-term Therapeutic Strategy for Prevention. American journal of ophthalmology. 2014;158:5–11. e12. [Europe PMC free article] [Abstract] [Google Scholar]
79. Chen Z, O'Shea JJ. Regulation of IL-17 production in human lymphocytes. Cytokine. 2008;41:71–78. [Abstract] [Google Scholar]
80. Gaffen SL. Recent advances in the IL-17 cytokine family. Current opinion in immunology. 2011;23:613–619. [Europe PMC free article] [Abstract] [Google Scholar]
81. Wei L, et al. Hypomethylation of the IL17RC promoter associates with age-related macular degeneration. Cell reports. 2012;2:1151–1158. [Europe PMC free article] [Abstract] [Google Scholar]
82. Oliver VF, et al. Hypomethylation of the IL17RC promoter in peripheral blood leukocytes is not a hallmark of age-related macular degeneration. Cell reports. 2013;5:1527–1535. [Europe PMC free article] [Abstract] [Google Scholar]
83. Handa JT. How does the macula protect itself from oxidative stress? Molecular aspects of medicine. 2012;33:418–435. [Europe PMC free article] [Abstract] [Google Scholar]
84. Gu X, et al. Carboxyethylpyrrole protein adducts and autoantibodies, biomarkers for age-related macular degeneration. The Journal of biological chemistry. 2003;278:42027–42035. [Abstract] [Google Scholar]
85. Hollyfield JG, et al. Oxidative damage-induced inflammation initiates age-related macular degeneration. Nature medicine. 2008;14:194–198. [Europe PMC free article] [Abstract] [Google Scholar]
86. Hollyfield JG, et al. A hapten generated from an oxidation fragment of docosahexaenoic acid is sufficient to initiate age-related macular degeneration. Molecular neurobiology. 2010;41:290–298. [Europe PMC free article] [Abstract] [Google Scholar]
87. Cruz-Guilloty F, et al. Infiltration of proinflammatory m1 macrophages into the outer retina precedes damage in a mouse model of age-related macular degeneration. International journal of inflammation. 2013;2013:503725. [Europe PMC free article] [Abstract] [Google Scholar]
88. Crabb JW, et al. Drusen proteome analysis: an approach to the etiology of age-related macular degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:14682–14687. [Europe PMC free article] [Abstract] [Google Scholar]
89. Gu J, et al. Proteomic and genomic biomarkers for age-related macular degeneration. Advances in experimental medicine and biology. 2010;664:411–417. [Abstract] [Google Scholar]
90. Aiello LP. Angiogenic pathways in diabetic retinopathy. The New England journal of medicine. 2005;353:839–841. [Abstract] [Google Scholar]
91. Connor KM, et al. Quantification of oxygen-induced retinopathy in the mouse: a model of vessel loss, vessel regrowth and pathological angiogenesis. Nature protocols. 2009;4:1565–1573. [Europe PMC free article] [Abstract] [Google Scholar]
92. Lambert V, et al. Laser-induced choroidal neovascularization model to study age-related macular degeneration in mice. Nature protocols. 2013;8:2197–2211. [Abstract] [Google Scholar]
93. Danis R, et al. Pazopanib eye drops: a randomised trial in neovascular age-related macular degeneration. The British journal of ophthalmology. 2014;98:172–178. [Abstract] [Google Scholar]
94. Xue Q, et al. Palomid 529, a novel small-molecule drug, is a TORC1/TORC2 inhibitor that reduces tumor growth, tumor angiogenesis, and vascular permeability. Cancer research. 2008;68:9551–9557. [Europe PMC free article] [Abstract] [Google Scholar]
95. Ciulla TA, et al. Squalamine lactate reduces choroidal neovascularization in a laser-injury model in the rat. Retina. 2003;23:808–814. [Abstract] [Google Scholar]
96. Smith AC, et al. Safety of Ad(GV)PEDF.11D administered by subtenon injection following laser disruption of the Bruch's membrane in cynomolgus monkey eyes. Investigative ophthalmology & visual science. 2003;44:U89–U89. [Google Scholar]
97. Russell SR, et al. Anecortave acetate for the treatment of exudative age-related macular degeneration - A review of clinical outcomes. Surv Ophthalmol. 2007;52:S79–S90. [Abstract] [Google Scholar]
98. Kaiser PK. Emerging therapies for neovascular age-related macular degeneration: drugs in the pipeline. Ophthalmology. 2013;120:S11–15. [Abstract] [Google Scholar]
99. Dalal M, et al. Subconjunctival Palomid 529 in the treatment of neovascular age-related macular degeneration. Graefe's archive for clinical and experimental ophthalmology = Albrecht von Graefes Archiv fur klinische und experimentelle Ophthalmologie. 2013;251:2705–2709. [Europe PMC free article] [Abstract] [Google Scholar]
100. Doukas J, et al. Topical administration of a multi-targeted kinase inhibitor suppresses choroidal neovascularization and retinal edema. J Cell Physiol. 2008;216:29–37. [Europe PMC free article] [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/2946280
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/2946280

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1016/j.molmed.2014.10.005

Supporting
Mentioning
Contrasting
3
88
0

Article citations


Go to all (77) article citations

Funding 


Funders who supported this work.

NEI NIH HHS (4)

NIAID NIH HHS (2)