Molecular Vision 2004; 10:964-972 <http://www.molvis.org/molvis/v10/a115/>
Received 9 March 2004 | Accepted 20 December 2004 | Published 22 December 2004
Download
Reprint


Systemic rapamycin inhibits retinal and choroidal neovascularization in mice

Nadine S. Dejneka, Akiko M. Kuroki, Joshua Fosnot, Waixing Tang, Michael J. Tolentino, Jean Bennett
 
 

F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA

Correspondence to: Jean Bennett, 310 Stellar-Chance Labs, 422 Curie Boulevard, Philadelphia, PA 19104-6069; Phone: (215) 898-0915; FAX: (215) 573-7155; email: jebennet@mail.med.upenn.edu


Abstract

Purpose: Rapamycin exhibits significant antitumor/antiangiogenic activity that is coupled with a decrease in vascular endothelial growth factor (VEGF) production and a reduction in the response of vascular endothelial cells to stimulation by VEGF. VEGF plays a significant role in neovascular pathologies of the eye, thus we tested the possibility of using rapamycin to inhibit retinal and choroidal neovascularization (CNV).

Methods: CNV was induced in adult mice with laser photocoagulation. Retinal neovascularization was induced using the retinopathy of prematurity (ROP) hyperoxia/hypoxia model. Experimental animals received intraperitoneal (ip) injections of rapamycin (2 mg/kg/day or 4 mg/kg/day) for 1-2 weeks. Controls were not treated or received ip injections of phosphate buffered saline (PBS). Eyes were analyzed histologically for evidence of CNV or retinal neovascularization. ROP eyes were further analyzed for changes in VEGF and VEGF receptor (Flt-1 and Flk-1) protein content following rapamycin treatment.

Results: Rapamycin significantly reduced the extent of neovascularization in both the CNV and the ROP model. Immunohistochemical staining of treated and untreated ROP retina did not reveal a significant reduction in levels of VEGF protein or its receptors. Immunostaining for Flt-1 increased, while no obvious changes in Flk-1 were observed. Quantitative analysis of total protein via enzyme linked immunosorbent assay (ELISA) confirmed an increase in Flt-1 and VEGF, following drug treatment, with no effect on Flk-1.

Conclusions: These results suggest rapamycin may provide an effective new treatment for ocular neovascularization.


Introduction

Age related macular degeneration (AMD) and retinopathy of prematurity (ROP) are characterized by aberrant ocular neovascularization that often results in severe, irreversible visual impairment. AMD is primarily responsible for vision loss in people over the age of 65 [1-3]. In the exudative (wet) form, choroidal blood vessels grow through Bruch's membrane into the subretinal space, resulting in accumulation of blood beneath the retina. As a result, retinal detachment and disciform scarring occur [4]. In contrast, ROP is characterized by retinal neovascularization that results from the exposure of the incompletely vascularized retina to high oxygen levels followed by a return to normoxic conditions, resulting in a relative hypoxic state [5]. This disease is a major cause of blindness in children in the developed world, and premature infants born less than 36 weeks gestation (<1250 g), requiring supplemental oxygen, are at greatest risk [6].

Mounting evidence strongly suggests that vascular endothelial growth factor (VEGF) plays a key role in these neovascular processes, and its inhibition can suppress the growth of abnormal blood vessels [7-12]. An upregulation of VEGF message in the ROP retina is well documented and its inhibition attenuates the observed retinal neovascularization [7-10]. In addition, VEGF has been associated with choroidal neovascular membranes, in RPE and maculae with AMD and in non-human primate and rodent laser models of choroidal neovascularization (CNV) [13-15]. Furthermore, subretinal injection of viral vectors encoding VEGF result in animal models of CNV [12,16,17].

Rapamycin, a macrolide fungicide with potent antimicrobial and immunosuppressive activities, possesses significant anti-tumor and anti-angiogenic properties [18,19]. The anti-angiogenic properties are associated with a decrease in VEGF production and a reduction in the response of vascular endothelial cells to stimulation by VEGF [19]. The mechanism by which rapamycin elicits its effects is not yet clear. Binding of rapamycin to its intracellular target, the immunophilin FKBP12, results in the inhibition of a kinase, termed the mammalian target of rapamycin (mTOR) [18]. mTOR, which is normally activated in response to growth signaling through the PI3K/Akt pathway, plays an important role in mitogenic cell signaling, affecting several biochemical processes that are critical in cell cycle regulation including translation initiation, DNA replication and protein synthesis [20]. Blockage of mTOR function inhibits PI3K/Akt signaling and results in cell cycle arrest [18].

In vitro studies in human smooth muscle and rodent endothelial cells indicate that mTOR inhibition by rapamycin specifically abrogates hypoxia triggered proliferation and angiogenesis [21]. Moreover, mTOR is an upstream activator of hypoxia inducible factor 1a (HIF-1a), a transcription factor that mediates the transcriptional regulation of VEGF [22,23]. In hypoxic cells rapamycin can interfere with HIF-1a activation by increasing the rate of its degradation [22]. Based on these findings, we tested the possibility of using rapamycin to inhibit neovascularization in a model of laser induced choroidal and ischemia induced retinal neovascularization, representing AMD [24] and ROP [25], respectively.


Methods

Animals

All experiments were conducted in accordance with the Animal Care and Use Committee and The Association for Research in Vision and Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research.

Laser induced model of CNV

Adult female C57Bl/6 mice (n=27) were anesthetized with avertin (2,2,2-tribromoethanol; 0.5 mg/g intraperitoneally (ip)) and pupils were dilated with 1% tropicamide. Laser photocoagulation was performed using a diode laser photocoagulator (OcuLight Six; IRIS Medical, Mountain View, CA) and a slit lamp system with a cover slip as a contact lens. Laser photocoagulation (130 mW, 75 μm spot size, 0.1 s duration) was applied to the 9, 12, and 3 o'clock positions, 2 to 3 disk diameters from the optic nerve of each eye. The rupture of Bruch's membrane was identified by a bubble formation (which occurred immediately) at the site of photocoagulation. Laser spots that did not result in the rupture of Bruch's membrane were excluded from the studies. Two weeks post-treatment, the size of CNV was measured using a flat mount technique, as previously described [11]. Briefly, mice were anesthetized and perfused through the left ventricle with a 30 gauge needle with 1 ml of 50 mg/ml fluorescein labeled dextran (2x106 average molecular weight; Sigma, St. Louis, MO). The eyes were enucleated and fixed in 10% formalin for 24 h. Choroidal flat mounts were made and analyzed by fluorescent microscopy using a Leica DMR microscope (Leica Microsystems, Wetzler, Germany) equipped with epifluorescence illumination. Openlab software (Improvision, Boston MA) was used to measure the magnitude of the hyperfluorescent areas corresponding to choroidal neovascularization. In addition, some eyes were processed for histology. Eyes were enucleated, fixed in 4% paraformaldehyde overnight, cryopreserved and 10 μm sections were obtained on a Reichert-Jung Cryocut 1800 (Leica Microsystems). Sections were subsequently stained with hematoxylin and eosin.

Hypoxia induced model of ROP

We used a murine model of ROP that was described previously and in which retinal neovascularization is induced reproducibly 5-9 days after return to room air [25]. Sixty-five ROP mice were created for 4 different experiments. Briefly, post-natal day (P) 7 C57Bl/6 pups were placed in a hyperoxic chamber (75% oxygen) for 5 days. On P12, the mice were removed from the chamber and placed in room air. A subset of animals was sacrificed on day P19, and the eyes were fixed in 4% paraformaldehyde, embedded in paraffin, sectioned serially (5 μm) and stained with hematoxylin and eosin. An individual masked to the treatment groups examined the sections for evidence of neovascularization via light microscopy. Neovascularization was quantified by counting the number of retinal vascular endothelial cell nuclei anterior to the inner limiting membrane (ILM). Counts were performed in transverse sections through the center of the eye (and the optic nerve head; see Figure 1D). Six to seventeen sections/eye were counted and counts were averaged.

In vivo rapamycin studies

Mice underwent laser photocoagulation as described above. Experimental groups received 4 mg/kg/day (n=9) or 2 mg/kg/day (n=9) of Rapamune (sirolimus; Wyeth Laboratories, Philadelphia, PA) ip starting the day of laser photocoagulation and then daily for 2 weeks. The control group (n=9) received PBS ip using the same schedule. The area of CNV was quantified as described above. ROP mice were removed from the hyperoxic chamber on P12. Animals subsequently received ip rapamycin injections for 7 days (2 mg/kg/day or 4 mg/kg/day). Pups were sacrificed at different time points. Eyes were enucleated and processed for paraffin (see above) or frozen and analyzed for levels of VEGF, Flt-1 and Flk-1 (see below). Control animals included: 1. age matched animals that developed normally in room air; 2. age matched animals that developed normally in room air and received rapamycin injections; 3. age matched animals that were exposed to hyperoxia and then returned to room air but did not receive rapamycin treatments. Experiments were repeated two times.

VEGF, Flt-1, and Flk-1 immunohistochemistry

Immunostaining was performed on frozen sections. Briefly, animals were euthanized and eyes were fixed in 4% paraformaldehyde. Following cryopreservation, 10 μm sections were obtained and blocked in PBS containing 1% horse serum, 1% BSA and 0.05% Triton X-100 in PBS for 40 min. Sections were subsequently treated with primary antibody directed against VEGF (VEGF A-20 rabbit polyclonal IgG, 1:100 dilution; Santa Cruz Biotechnology, Inc., Santa Cruz, CA), Flt-1 (Flt-1 H-225 rabbit polyclonal IgG, 1:100 dilution; Santa Cruz Biotechnology, Inc.) or Flk-1 (A-3 mouse monoclonal IgG, 1:50 dilution; Santa Cruz Biotechnology, Inc.) prepared in PBS containing 1% horse serum, 1% BSA and 0.3% Triton X-100 (1 h room temp). Sections were washed with PBS and then incubated in secondary antibody (Alexa Fluor 488 goat anti-rabbit IgG (H+L), 1:100 dilution, Molecular Probes, Eugene, OR or Alexa Fluor 488 goat anti-mouse IgG (H+L), 1:100 dilution, Molecular Probes) prepared in PBS containing 1% horse serum, 1% BSA and 0.3% Triton X-100 (1 h at ambient room temperature). Slides were washed with PBS, coverslipped with mounting media containing DAPI and evaluated with a Leica DME microscope (Leica Microsystems, Inc.) equipped with epifluorescence. Images were captured with a Hamamatsu digital camera and Openlab 2.2 image analysis software (Improvision, Inc.). To test the specificity of antibody, control sections were stained simultaneously following the same procedures; however, primary antibody was omitted. Alternatively, an irrelevant primary antibody was added.

VEGF, Flt-1, and Flk-1 enzyme linked immunosorbent assays

Upon enucleation, conjunctiva was removed, and eyes were stored at -80 °C. Samples were homogenized in a buffer containing 10 mM KH2PO4, 150 mM NaCl, 200 mM sucrose, 10 mM EDTA, 10 mg/ml leupeptin, 10 mg/ml pepstatin A and 1 mM PMSF. Samples were subsequently centrifuged at 6,500 rpm for 7 min. The supernatants were analyzed for protein concentrations via the Bio-Rad Protein Assay Reagent (Bio-Rad Laboratories, Hercules, CA). Samples were diluted to a final concentration of 2 mg/ml. Levels of VEGF, Flt-1, and Flk-1 were determined using the Quantikine M Murine VEGF, sVEGF R1, and sVEGF R2 Immunoassay Kits, respectively (R&D Systems Inc., Minneapolis, MN). Five to 14 tissue samples were analyzed for each group and time point.

Statistical analysis

The statistical significance of differential findings between experimental and control groups was determined by an unpaired Student's t-test and considered significant if two tailed p values were <0.05.


Results

Effect of rapamycin administration on CNV

Animals that received rapamycin ip for 2 weeks did not manifest any significant side effects such as weight loss, severe infection, or death due to the drug, and maintained normal appetite and activity. The laser induced model yielded a mean area of CNV equal to 0.055±0.006 (SEM) mm2 in the control (no rapamycin) group, 0.039±0.004 mm2 in the 2 mg/kg group, and 0.033±0.003 mm2 in the 4 mg/kg rapamycin group, as indicated by analysis of choroidal flat mounts (Figure 2A,B,E). This translated into a 29.8% decrease in CNV area in the low dose group and a 40% decrease in the high dose group, with respect to the control, that was statistically significant (p<0.05 and <0.001, respectively). Representative cryopreserved histological sections show the difference in area of neovascularization before and after treatment (Figure 2C,D).

Effect of rapamycin administration on retinal neovascularization

Mice with oxygen induced ischemic retinopathy received ip injections of rapamycin (2 mg/kg or 4 mg/kg) once daily between P12 and P18. Age matched normoxic animals also received drug treatment. Additional normoxic and ROP controls were maintained without therapeutic intervention. Rapamycin had no apparent systemic effects on the normoxic animals. Hypoxic animals exhibited weight loss for 3-4 days of treatment, but then slowly regained weight. The ROP model yielded 61.3±5.0 (SEM) vascular nuclei per 5 μm cross section, extending from the ILM into the vitreous on P19. This number was significantly reduced following daily rapamycin treatments (Figure 1). The 2 and 4 mg/kg groups averaged 16.9±2 (p<0.001) and 11.6±2 (p<0.001) nuclei, respectively. This data correlated with a 72.4% decrease in neovascular tuft extension into the vitreous with low dose rapamycin and an 81.1% reduction with the high dose.

VEGF, Flt-1, and Flk-1 immunohistochemistry

Immunohistochemistry was performed in order to assess whether rapamycin treatment affected patterns of distribution of VEGF protein and its receptors in the ROP retina (Figure 3). We observed VEGF protein throughout the retina, with highest levels in the ganglion cell layer (GCL), especially surrounding the neovascular tufts. VEGF levels were also high in the outer plexiform layer (OPL), inner plexiform layer (IPL). The two receptors showed expression in these regions. Similar patterns of expression were observed in the age matched normoxic controls (data not shown). Interestingly, high dose rapamycin increased Flt-1 labeling. No obvious changes were readily detectable in VEGF or Flk-1 labeling. Similar patterns of expression were seen with the low dose group (data not shown).

Quantitative analysis of VEGF, Flt-1, and Flk-1

In order to confirm our immunohistochemical observations, we used enzyme linked immunosorbent assays (ELISAs) to quantify VEGF, Flt-1 and Flk-1 levels in the eye. Conjunctiva was removed from all samples at the time of enucleation, as VEGF protein is normally found in this tissue in rodents and non-human primates [26]. Our data indicate rapamycin does not reduce levels of VEGF or its receptors in either the normal or ROP developing eye (Figure 4). Upon initial removal of animals from the hyperoxia chamber, VEGF protein levels were similar to those of untreated 12 day old controls (35.2±12.4 pg/ml and 43.4±3.6 pg/ml, respectively). However, VEGF concentrations in the treated mice increased significantly by day 15 (174.7±7.6 pg/ml) and remained elevated (Figure 4A). Rapamycin treatment resulted in comparably high VEGF levels on postnatal (P) day 15 (181.1±12.9 pg/ml), and these levels remained high through all the time points examined. No effect was seen on VEGF protein levels in eyes of animals raised in room air, where levels remain low and slowly decrease over time.

Flt-1 levels in ROP mice were also analogous to those of untreated controls on P12 (1762.5±155.4 pg/ml compared to 1695.6±175.9 pg/ml; Figure 4B). A gradual rise in receptor content is observed in the ROP group between P13-P17, but by P19 levels begin to decline. Rapamycin treatment increased Flt-1 levels in the ROP group and had no effect on the controls. Interestingly, we observed a significant elevation in Flk-1 following oxygen exposure (from 35.0±4.9 pg/ml to 11.8±5.4 pg/ml in age matched normoxic controls) that decreases to normal levels by P15 (16.2±1.8 pg/ml; Figure 4C). Rapamycin had no apparent effect on the decrease we observed, nor did it affect overall levels in developing eyes of animals maintained under normoxic conditions.


Discussion

In the present study, we demonstrated the efficacy of ip rapamycin for the treatment of CNV and retinal neovascularization in mice. Both pathologies have been associated with increased levels of VEGF [5-7,13-15]. Thus, we examined the effects of rapamycin on levels of VEGF and its receptors in the ROP retina. Studies in a murine tumor model demonstrated that administration of rapamycin reduced VEGF production and decreased the vascular endothelial cells' sensitivity to VEGF stimulation [19]. The murine VEGF gene is alternatively transcribed, resulting in three different isoforms (VEGF120, VEGF164, and VEGF188) that differ in molecular mass, solubility, receptor binding, and function [27]. While we are not aware of any reports that specifically link rapamycin's effect to any one single isoform, it is known that during pathological ischemia induced retinal neovascularization, absolute and relative expression levels of VEGF164 are increased, and it selectively induces inflammation and cellular immunity [28]. In addition, it has been identified as an important isoform in the pathogenesis of early diabetic retinopathy [29].

In vitro studies revealed rapamycin is capable of blocking the activation of HIF-1a, the transcription factor that induces the expression of VEGF and its receptor Flt-1 [22,23]. Curiously, our ELISA and immunohistochemical data revealed that VEGF levels are not reduced in the ROP model. Instead, these levels increase following treatment even though the extent of neovascularization is significantly reduced. Interestingly, Flt-1 protein levels also increase following treatment, suggesting that Flt-1 may play a role in minimizing the pathological changes in the ROP retina. In contrast, we did not observe any changes in Flk-1 levels after treatment with rapamycin.

It is not apparent by what mechanism rapamycin could increase Flt-1. Flt-1 has been localized to neonatal blood vessels, while Flk-1 is selectively localized outside the vasculature in the neonatal retina until P12, when it is also seen on retinal vessels [30]. While Flt-1 message is upregulated between P3 and P26, there are no significant changes in Flk-1 mRNA expression during this time. The individual involvement of Flt-1 and Flk-1 in oxygen induced retinal vessel degeneration has been examined in vivo using specific ligands that selectively bind each of the receptors [30]. Specific activation of Flt-1 with placental growth factor-1 (PlGF-1) protects against oxygen induced vessel loss without stimulating vascular proliferation and neovascularization; however, VEGF-E, which selectively activates Flk-1, does not prevent oxygen induced vaso-obliteration. These findings strongly suggest that stimulating Flt-1 by PlGF-1 may prove to be an attractive strategy for preventing ROP. Transforming growth factor b1 (TGF-b1) also induces Flt-1 expression in endothelial cells, and co administration of TGF-b1 and PlGF-1 further increase protection from hyperoxia induced degeneration [31].

It has been suggested that Flt-1 can act as a "decoy" receptor to prevent the binding of VEGF to Flk-1, the major mediator of the mitogenic, angiogenic and permeability enhancing effects of VEGF [32]. Studies with a soluble form of Flt-1 show it causes both the sequestration of VEGF and forms inactive heterodimers with membrane spanning isoforms of Flt-1 and Flk-1 [33,34]. Indeed, it has been demonstrated that viral mediated delivery of a soluble form of Flt-1 results in an inhibition of corneal, choroidal and retinal neovascularization [32,35-37].

Rapamycin may also be exerting its effects through additional non-VEGF related pathways. One possibility is that rapamycin is blocking the production of other powerful pro-angiogenic agents, as high levels of VEGF are not sufficient to induce the neovascularization observed in ROP and CNV, and VEGF inhibitors only partially block the neovascular response [38,39]. Hypoxia can stimulate gene expression by at least three different mechanisms including gene transcription, regulation at the translational level and mRNA stabilization, and mTOR, the target of rapamycin, has effects on all of these pathways [18-20,40]. Factors such as basic fibroblast growth factor (bFGF), platelet derived growth factor B (PDGF-B), nitric oxide synthase and angiopoietin are upregulated by hypoxia [21]. In a study that examined the effects of VEGF, bFGF, and PDGF-B on reconstituted basement membrane matrix experiments, it was found that only bFGF and PDGF-B could exert a secondary sprouting effect alone [41]. Alternatively, rapamycin may be directly or indirectly stimulating the production of one or more anti-angiogenic factors that are responsible for the dramatic effects we observed. Anti-angiogenic molecules such as pigment epithelium derived factor (PEDF), tissue inhibitor of metalloproteinase-3 (TIMP3) and endostatin have all shown efficacy in inhibiting both CNV and retinal neovascularization [42].

The mechanism by which rapamycin elicits its anti-angiogenic properties in the eye is not clear. Nevertheless, this study provides evidence that rapamycin inhibits ocular neovascularization in two different models. These results warrant further testing to determine the stability of the effect and the effective dose range of the drug. We selected our dosages based on a previous report that indicated 1.5 mg/kg/day rapamycin (about 40 ng/ml blood trough level) was effective in inhibiting tumor angiogenisis and decreasing VEGF production [19]. Adult transplant patients on a rapamycin regimen maintain a dose of 2 mg/day following an initial one-time dose of 6 mg [43], with recommended blood trough levels of 4-20 ng/ml [44]. The antiangiogenic effects we observed did not significantly differ between the two concentrations of drug we tested, and it may be possible to achieve similar effects with substantially lower doses. It may also be possible to deliver rapamycin locally to the eye via topical or subretinal injection in order to limit side effects that might appear after systemic exposure. These possibilities and the relative safety of this approach, with respect to the eye and the rest of the body, are under investigation.


Acknowledgements

Supported by the Juvenile Diabetes Research Foundation, R01 EY10820 and EY12156, the Foundation Fighting Blindness, William and Mary Greve International Research Scholar Award from Research to Prevent Blindness, Inc., the Ruth and Milton Steinbach Fund (JB), the Paul and Evanina Mackall Trust, and the F. M. Kirby Foundation.


References

1. Allikmets R, Shroyer NF, Singh N, Seddon JM, Lewis RA, Bernstein PS, Peiffer A, Zabriskie NA, Li Y, Hutchinson A, Dean M, Lupski JR, Leppert M. Mutation of the Stargardt disease gene (ABCR) in age-related macular degeneration. Science 1997; 277:1805-7.

2. Ferris FL 3rd, Fine SL, Hyman L. Age-related macular degeneration and blindness due to neovascular maculopathy. Arch Ophthalmol 1984; 102:1640-2.

3. Argon laser photocoagulation for neovascular maculopathy. Five-year results from randomized clinical trials. Macular Photocoagulation Study Group. Arch Ophthalmol 1991; 109:1109-14. Erratum in: Arch Ophthalmol 1992; 110:761.

4. Green WR. Histopathology of age-related macular degeneration. Mol Vis 1999; 5:27 <http://www.molvis.org/molvis/v5/a27/>.

5. Hellstrom A, Perruzzi C, Ju M, Engstrom E, Hard AL, Liu JL, Albertsson-Wikland K, Carlsson B, Niklasson A, Sjodell L, LeRoith D, Senger DR, Smith LE. Low IGF-I suppresses VEGF-survival signaling in retinal endothelial cells: direct correlation with clinical retinopathy of prematurity. Proc Natl Acad Sci U S A 2001; 98:5804-8.

6. Friedman NJ, Pineda R, Kaiser PK. The Massachusetts Eye and Ear Infirmary illustrated manual of ophthalmology. Philadelphia: WB Saunders; 1998.

7. Pierce EA, Avery RL, Foley ED, Aiello LP, Smith LE. Vascular endothelial growth factor/vascular permeability factor expression in a mouse model of retinal neovascularization. Proc Natl Acad Sci U S A 1995; 92:905-9.

8. Aiello LP, Pierce EA, Foley ED, Takagi H, Chen H, Riddle L, Ferrara N, King GL, Smith LE. Suppression of retinal neovascularization in vivo by inhibition of vascular endothelial growth factor (VEGF) using soluble VEGF-receptor chimeric proteins. Proc Natl Acad Sci U S A 1995; 92:10457-61.

9. Robinson GS, Pierce EA, Rook SL, Foley E, Webb R, Smith LE. Oligodeoxynucleotides inhibit retinal neovascularization in a murine model of proliferative retinopathy. Proc Natl Acad Sci U S A 1996; 93:4851-6.

10. Bainbridge JW, Jia H, Bagherzadeh A, Selwood D, Ali RR, Zachary I. A peptide encoded by exon 6 of VEGF (EG3306) inhibits VEGF-induced angiogenesis in vitro and ischaemic retinal neovascularisation in vivo. Biochem Biophys Res Commun 2003; 302:793-9.

11. Reich SJ, Fosnot J, Kuroki A, Tang W, Yang X, Maguire AM, Bennett J, Tolentino MJ. Small interfering RNA (siRNA) targeting VEGF effectively inhibits ocular neovascularization in a mouse model. Mol Vis 2003; 9:210-6 <http://www.molvis.org/molvis/v9/a31/>.

12. Wang F, Rendahl KG, Manning WC, Quiroz D, Coyne M, Miller SS. AAV-mediated expression of vascular endothelial growth factor induces choroidal neovascularization in rat. Invest Ophthalmol Vis Sci 2003; 44:781-90.

13. Kliffen M, Sharma HS, Mooy CM, Kerkvliet S, de Jong PT. Increased expression of angiogenic growth factors in age-related maculopathy. Br J Ophthalmol 1997; 81:154-62.

14. Ryan SJ. Subretinal neovascularization. Natural history of an experimental model. Arch Ophthalmol 1982; 100:1804-9.

15. Yi X, Ogata N, Komada M, Yamamoto C, Takahashi K, Omori K, Uyama M. Vascular endothelial growth factor expression in choroidal neovascularization in rats. Graefes Arch Clin Exp Ophthalmol 1997; 235:313-9.

16. Spilsbury K, Garrett KL, Shen WY, Constable IJ, Rakoczy PE. Overexpression of vascular endothelial growth factor (VEGF) in the retinal pigment epithelium leads to the development of choroidal neovascularization. Am J Pathol 2000; 157:135-44. Erratum in: Am J Pathol 2000; 157:1413.

17. Baffi J, Byrnes G, Chan CC, Csaky KG. Choroidal neovascularization in the rat induced by adenovirus mediated expression of vascular endothelial growth factor. Invest Ophthalmol Vis Sci 2000; 41:3582-9.

18. Sausville EA, Elsayed Y, Monga M, Kim G. Signal transduction--directed cancer treatments. Annu Rev Pharmacol Toxicol 2003; 43:199-231.

19. Guba M, von Breitenbuch P, Steinbauer M, Koehl G, Flegel S, Hornung M, Bruns CJ, Zuelke C, Farkas S, Anthuber M, Jauch KW, Geissler EK. Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nat Med 2002; 8:128-35.

20. Sehgal SN. Rapamune (RAPA, rapamycin, sirolimus): mechanism of action immunosuppressive effect results from blockade of signal transduction and inhibition of cell cycle progression. Clin Biochem 1998; 31:335-40.

21. Humar R, Kiefer FN, Berns H, Resink TJ, Battegay EJ. Hypoxia enhances vascular cell proliferation and angiogenesis in vitro via rapamycin (mTOR)-dependent signaling. FASEB J 2002; 16:771-80.

22. Hudson CC, Liu M, Chiang GG, Otterness DM, Loomis DC, Kaper F, Giaccia AJ, Abraham RT. Regulation of hypoxia-inducible factor 1alpha expression and function by the mammalian target of rapamycin. Mol Cell Biol 2002; 22:7004-14.

23. Gerber HP, Condorelli F, Park J, Ferrara N. Differential transcriptional regulation of the two vascular endothelial growth factor receptor genes. Flt-1, but not Flk-1/KDR, is up-regulated by hypoxia. J Biol Chem 1997; 272:23659-67.

24. Kwak N, Okamoto N, Wood JM, Campochiaro PA. VEGF is major stimulator in model of choroidal neovascularization. Invest Ophthalmol Vis Sci 2000; 41:3158-64.

25. Smith LE, Wesolowski E, McLellan A, Kostyk SK, D'Amato R, Sullivan R, D'Amore PA. Oxygen-induced retinopathy in the mouse. Invest Ophthalmol Vis Sci 1994; 35:101-11.

26. Kim I, Ryan AM, Rohan R, Amano S, Agular S, Miller JW, Adamis AP. Constitutive expression of VEGF, VEGFR-1, and VEGFR-2 in normal eyes. Invest Ophthalmol Vis Sci 1999; 40:2115-21. Erratum in: Invest Ophthalmol Vis Sci 2000; 41:368.

27. Stalmans I, Ng YS, Rohan R, Fruttiger M, Bouche A, Yuce A, Fujisawa H, Hermans B, Shani M, Jansen S, Hicklin D, Anderson DJ, Gardiner T, Hammes HP, Moons L, Dewerchin M, Collen D, Carmeliet P, D'Amore PA. Arteriolar and venular patterning in retinas of mice selectively expressing VEGF isoforms. J Clin Invest 2002; 109:327-36.

28. Ishida S, Usui T, Yamashiro K, Kaji Y, Amano S, Ogura Y, Hida T, Oguchi Y, Ambati J, Miller JW, Gragoudas ES, Ng YS, D'Amore PA, Shima DT, Adamis AP. VEGF164-mediated inflammation is required for pathological, but not physiological, ischemia-induced retinal neovascularization. J Exp Med 2003; 198:483-9.

29. Ishida S, Usui T, Yamashiro K, Kaji Y, Ahmed E, Carrasquillo KG, Amano S, Hida T, Oguchi Y, Adamis AP. VEGF164 is proinflammatory in the diabetic retina. Invest Ophthalmol Vis Sci 2003; 44:2155-62.

30. Shih SC, Ju M, Liu N, Smith LE. Selective stimulation of VEGFR-1 prevents oxygen-induced retinal vascular degeneration in retinopathy of prematurity. J Clin Invest 2003; 112:50-7.

31. Shih SC, Ju M, Liu N, Mo JR, Ney JJ, Smith LE. Transforming growth factor beta1 induction of vascular endothelial growth factor receptor 1: mechanism of pericyte-induced vascular survival in vivo. Proc Natl Acad Sci U S A 2003; 100:15859-64.

32. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003; 9:669-76.

33. Kendall RL, Thomas KA. Inhibition of vascular endothelial cell growth factor activity by an endogenously encoded soluble receptor. Proc Natl Acad Sci U S A 1993; 90:10705-9.

34. Kendall RL, Wang G, Thomas KA. Identification of a natural soluble form of the vascular endothelial growth factor receptor, FLT-1, and its heterodimerization with KDR. Biochem Biophys Res Commun 1996; 226:324-8.

35. Lai CM, Brankov M, Zaknich T, Lai YK, Shen WY, Constable IJ, Kovesdi I, Rakoczy PE. Inhibition of angiogenesis by adenovirus-mediated sFlt-1 expression in a rat model of corneal neovascularization. Hum Gene Ther 2001; 12:1299-310.

36. Lai YK, Shen WY, Brankov M, Lai CM, Constable IJ, Rakoczy PE. Potential long-term inhibition of ocular neovascularisation by recombinant adeno-associated virus-mediated secretion gene therapy. Gene Ther 2002; 9:804-13.

37. Bainbridge JW, Mistry A, De Alwis M, Paleolog E, Baker A, Thrasher AJ, Ali RR. Inhibition of retinal neovascularisation by gene transfer of soluble VEGF receptor sFlt-1. Gene Ther 2002; 9:320-6.

38. Okamoto N, Tobe T, Hackett SF, Ozaki H, Vinores MA, LaRochelle W, Zack DJ, Campochiaro PA. Transgenic mice with increased expression of vascular endothelial growth factor in the retina: a new model of intraretinal and subretinal neovascularization. Am J Pathol 1997; 151:281-91.

39. Schwesinger C, Yee C, Rohan RM, Joussen AM, Fernandez A, Meyer TN, Poulaki V, Ma JJ, Redmond TM, Liu S, Adamis AP, D'Amato RJ. Intrachoroidal neovascularization in transgenic mice overexpressing vascular endothelial growth factor in the retinal pigment epithelium. Am J Pathol 2001; 158:1161-72.

40. Treins C, Giorgetti-Peraldi S, Murdaca J, Semenza GL, Van Obberghen E. Insulin stimulates hypoxia-inducible factor 1 through a phosphatidylinositol 3-kinase/target of rapamycin-dependent signaling pathway. J Biol Chem 2002; 277:27975-81.

41. Castellon R, Hamdi HK, Sacerio I, Aoki AM, Kenney MC, Ljubimov AV. Effects of angiogenic growth factor combinations on retinal endothelial cells. Exp Eye Res 2002; 74:523-35.

42. Auricchio A, Behling KC, Maguire AM, O'Connor EM, Bennett J, Wilson JM, Tolentino MJ. Inhibition of retinal neovascularization by intraocular viral-mediated delivery of anti-angiogenic agents. Mol Ther 2002; 6:490-4.

43. Machado PG, Felipe CR, Hanzawa NM, Park SI, Garcia R, Alfieri F, Franco M, Silva HT Jr, Medina-Pestana JO. An open-label randomized trial of the safety and efficacy of sirolimus vs. azathioprine in living related renal allograft recipients receiving cyclosporine and prednisone combination. Clin Transplant 2004; 18:28-38.

44. Kahan BD, Napoli KL, Kelly PA, Podbielski J, Hussein I, Urbauer DL, Katz SH, Van Buren CT. Therapeutic drug monitoring of sirolimus: correlations with efficacy and toxicity. Clin Transplant 2000; 14:97-109.


Dejneka, Mol Vis 2004; 10:964-972 <http://www.molvis.org/molvis/v10/a115/>
©2004 Molecular Vision <http://www.molvis.org/molvis/>
ISSN 1090-0535