Strain-Dependent Increases in Retinal Inflammatory and Photoreceptor FGF-2 Expression in Streptozotocin-Induced Diabetic Rats

Stefanie J. Kirwin,1 Suzanne T. Kanaly,2 Noelle A. Linke,2 and Jeffrey L. Edelman1

1 PURPOSE. Inflammation is thought to play a role in disease progression is by tightly controlling blood glucose levels. progression and vision loss in diabetic retinopathy (DR). How- Despite significant morbidity and effect on quality of life, there ever, the level of inflammation and the role of cytokines and is currently no approved pharmacotherapy for this sight-threat- growth factors in the early stages of this disease are poorly ening disease.1,3 understood. Streptozotocin (STZ)-induced hyperglycemia in In the past, DR was considered primarily a vascular disease. rats is widely used as a model of diabetic retinopathy, and It has been described as and is still classified by abnormalities therefore this model was used to better define the inflamma- of the retinal vasculature that progress from hyperpermeability tory response and the impact of the genetic background. of the vessels, to capillary nonperfusion and hemorrhaging, METHODS. The expression of a panel of 57 inflammatory pro- followed by neovascularization of the retina. Macular edema teins and growth factors in the retina of three rat strains was can develop independently of other changes and poses a seri- compared by using a highly sensitive flow cytometry–based ous threat to central vision.1,2 However, there is mounting assay. Hyperglycemia was induced in Brown Norway (BN), evidence that neuropathy develops before and independent of Long-Evans (LE), and Sprague-Dawley (SD) rats, and vascular complications and contributes to the disease.4 Inflam- expression in the retina was measured 4 weeks and 3 months matory molecules such as cytokines can be found in the retina later. of diabetic patients,5 although it is presently unclear whether inflammation contributes to DR or is secondary to it. RESULTS. The data revealed a subtle, but reproducible, inflam- matory response in the retina of SD, but not in those of BN or Streptozotocin (STZ)-induced diabetes in the rat is com- LE, rats. Upregulation of fibroblast growth factor (FGF)-2 in the monly used as an experimental model of DR. The STZ rat photoreceptor nuclear layer coincided with the inflammatory model mimics the human disease by inducing hyperglycemia ␤ 6 response in SD rats and may constitute a neuroprotective after destruction of the -cells in the pancreas. Although there mechanism. Reduced expression of involved in the pho- are vascular changes in this model, the vasculopathy does not totransduction pathway indicates altered photoreceptor func- progress to neovascularization as is observed in humans. The tion. length of time that passes between the beginning of the hy- perglycemic insult and the development of symptoms is also a ONCLUSIONS Taken together, these data show that inflamma- C . challenge as are the number of different mechanisms that may tory changes in the diabetic rat retina are highly strain depen- cause damage.7 In addition, inflammatory changes are subtle dent, and SD rats exhibit low-level inflammation similar to that and therefore sometimes difficult to demonstrate reproducibly. observed in diabetic patients. Therefore, SD rats may be a good Further complicating matters is the fact that different rat strains model for the study of early inflammatory changes in human show different responses to hyperglycemia and other insults to diabetic retinopathy. (Invest Ophthalmol Vis Sci. 2009;50: the retina.8–10 5396–5404) DOI:10.1167/iovs.09-3474 In this study, we characterized the inflammatory changes in iabetic retinopathy (DR) is the number one cause of blind- the retina of diabetic rats, while exploring the effect of genetic Dness in working-age people in the United States today, and background, including pigmentation, on such changes. A its prevalence is expected to rise, with a projected increase in highly sensitive flow cytometry-based assay (Luminex Corp., the number of patients with type II diabetes. Currently, the Austin, TX) was used to screen for a large number of cytokines only available treatment for DR is laser photocoagulation, and and growth factors in the retina of diabetic Brown Norway although it is highly effective in preserving sight, it can have (BN), Long-Evans (LE), and Sprague-Dawley (SD) rats. These rat serious side effects.1 Furthermore, this treatment is efficacious strains were chosen due to their documented use in the STZ- only in proliferative DR and cannot restore vision already lost induced DR model and their various levels of pigmentation, to the disease.2 Most patients with diabetes develop retinopa- with SD being the least pigmented and BN the most pigmented thy at some point, and the only proven method of slowing its rat strain. To date LE and BN rats have been used mostly to study vascular abnormalities in diabetes such as increased leu- kostasis11–16 and blood–retina barrier breakdown.9,14,15,17–21 However, electrophysiological studies indicate functional def- From the Departments of 1Biological Sciences and 2Pathology, icits in the neural retina22–24 and results from studies on ␥-ami- Allergan, Inc., Irvine, California. 24,25 26 Supported by Allergan, Inc. nobutyric acid signaling and microarray data in LE as 27 Submitted for publication January 28, 2009; revised March 27 and well as Mu¨ller cell abnormalities in BN rats suggest that other May 6, 2009; accepted July 28, 2009. hallmarks of experimental DR occur and that these strains Disclosure: S.J. Kirwin, Allergan, Inc. (E, F); S.T. Kanaly, Aller- merit further investigation. gan, Inc. (E, F); N.A. Linke, Allergan, Inc. (E, F); J.L. Edelman, The data show that despite the absence of gross inflamma- Allergan, Inc. (E, F) tory infiltrates in the retina, markers of inflammation are The publication costs of this article were defrayed in part by page present in SD rats, but not the other two strains. Furthermore, charge payment. This article must therefore be marked “advertise- ment” in accordance with 18 U.S.C. §1734 solely to indicate this fact. the data show that the cytokine response coincides with up- Corresponding author: Stefanie J. Kirwin, Allergan, Inc., Biological regulation of fibroblast growth factor (FGF-2) in photorecep- Science, 2525 Dupont Drive, Irvine, CA 92612; tors, and expression analysis suggests impairment of [email protected]. photoreceptor function.

Investigative Ophthalmology & Visual Science, November 2009, Vol. 50, No. 11 5396 Copyright © Association for Research in Vision and Ophthalmology

Downloaded from iovs.arvojournals.org on 10/01/2021 IOVS, November 2009, Vol. 50, No. 11 Inflammation in the Retina of Diabetic Rats 5397

MATERIALS AND METHODS lerica, CA) at 1:200 dilution for 60 minutes. After they were washed with PBST, the tissues were reacted with biotinylated donkey anti- Animals and Induction of Diabetes mouse antibody (Jackson ImmunoResearch Laboratories, West Grove, PA) for 20 minutes, washed with PBST, and incubated for 20 minutes Diabetes was induced in male BN, LE, and SD rats (Charles River with horseradish-peroxidase streptavidin (Covance, Dedham, MA) and Laboratories, Wilmington, MA) weighing 250 to 300 g (BN: 244 Ϯ 19 g; rinsed in PBST. Signal was detected with AEC chromogen (Romulin; LE: 298 Ϯ 25 g; SD: 282 Ϯ 26 g) by intraperitoneal injection of 65 BioCare Medical, Concord, CA), then washed with PBST. Hematoxylin mg/kg bodyweight STZ in 0.09 M citrate buffer (pH 4.8; both Sigma- was applied for 5 minutes followed by a rinse with distilled water. Aldrich, St. Louis, MO). Age-matched control rats were injected with buffer only. At the time of injection, BN rats were moderately, but RNA Extraction and Real-Time PCR significantly (P Ͻ 0.05) smaller then LE or SD rats. Blood glucose was Retinas were removed and placed individually into tubes containing measured (Ascencia control system; Bayer, Tarrytown, NY), and rats 200 ␮L RNA stabilizer (RNAlater; Ambion, Austin, TX). After 24 hours with blood glucose levels above 220 mg/dL after 48 hours were at 4°C, excess fluid was removed and the samples were stored at deemed diabetic. Nondiabetic STZ-injected rats were reinjected with a Ϫ70°C until analysis. Frozen samples were placed into 1 mL lysis second dose on day 2.28–30 There was no significant difference in reagent (Qiazol; Qiagen, Valencia, CA) and homogenized (Tissue- weight or blood glucose levels 6 days after the initial injection between Tearor; Biospec, Bartlesville, OK). RNA was extracted (RNeasy Mini rats injected once and those receiving a second dose. No insulin was Kit; Qiagen) according to the manufacturers’ instructions. RNA quality supplied at any time. Occasionally, glucose levels exceeded the upper was then assessed (RNA 6000 Nano Kit; Agilent Technologies, Wald- limit of detection, and this unknown value was replaced by 600 mg/dL. bronn, Germany) and quantity determined by spectrophotometry. Re- Glucose spikes to these high levels were relatively rare and not limited verse transcription was performed on 1 ␮g of RNA using 25 mM MgCl , to any particular rat strain. Intraocular blood glucose was measured by 2 10 mM dNTP mixture, 25 U RNasin, and 15 U AMV reverse transcrip- making a 4- to 6-mm diagonal incision across the cornea, removing the tase in reverse transcription buffer (all Promega, Madison, WI) in 20 ␮L lens and then sampling the liquid in the eye cup using the capillary for 20 minutes at 42°C, followed by a 5-minute denaturing step at 95°C action of the control test stick (Ascencia; Bayer). The sample most and 5 minutes at 4°C. After reverse transcription, ddH O was added to likely contains a mixture of aqueous and vitreous humor. Animals were 2 a final volume of 200 ␮L. killed 4 weeks and 3 months after induction of diabetes. They were Real-time PCR was performed (7900HT Real-Time PCR System; Applied housed in plastic box cages in a specific pathogen-free, AALAC certi- Biosystems [ABI], Foster City, CA), with probe and primer sets (Taqman; ABI) fied facility on a 12-hour light/dark cycle. Room temperature was for Actb (ID: Rn00667869_m1), Arr3 (ID: Rn01424383_m1), Gnb3 maintained between 16°C and 23°C, average daily humidity between (Rn00516381_m1), Grk1 (ID: Rn00579470_m1), and Opn1mw (ID: 30% and 70%, and airflow between 10 and 30 air changes per hour. All Rn00585560_m1). All primers span exon junctions and therefore do animals were fed standard chow and treated according to the ARVO not recognize genomic DNA. Gene expression was normalized and Statement for the Use of Animals in Ophthalmic and Vision Research. converted to a linearized value by the following formula: unit ϭ Protein Analysis. Isolated neural retinas were snap frozen in ٙ (Ct Ϫ Ct ) ϫ 100.33 1.8 liquid nitrogen and samples were stored at Ϫ70°C until processing. actb gene x One retina each from three individual rats was combined for protein Statistical Analysis analysis. Protein was extracted by sonication of three pooled retinas in 300 ␮L Tris HCl (pH 7.5; Mediatech, Inc. Herndon, VA) containing Experiments analyzing retinal proteins were repeated at least three Complete, Mini protease inhibitor cocktail (Roche, Mannheim, Ger- times, each group containing three rats, making data the average of at many). Serum was collected by letting blood clot for 1 hour at room least nine rats from at least three independent experiments. Signifi- temperature, followed by a 10-minute centrifugation at 10,000g. Pro- cance was determined by Student’s t-test, unless otherwise noted. P Ͻ tein concentration was then determined (RodentMA Antigen 1.6 panel; 0.05 was considered significant. Rules Based Medicine Austin, TX). The assay uses mouse antibodies that cross-react with rat cytokines.31,32 Briefly, samples are incubated with microsphere multiplexes, which contain covalently linked assay RESULTS specific capture reagents, for 1 hour at room temperature before STZ-Induced Hyperglycemia in BN, LE, biotinylated reporter antibodies are added for an additional hour at room temperature. After incubation for 1 hour with streptavidin-phy- and SD Rats coerythrin solution and vacuum filtration, the samples are diluted in Diabetes was induced with STZ in three rat strains: BN, LE, and matrix buffer and analyzed with a multiplex bio-assay analyzer (100 IS SD. Over the 3-month observation period, average blood glu- System; Luminex Corp.). cose levels ranged from 330 to 542 mg/dL in the three rat Histopathology and Immunohistochemistry. Rats were strains (Fig. 1A), similar to previously published results in this killed after 3 months of diabetes, and the eyes were immediately fixed model.34–36 In contrast, vehicle-treated rats exhibited tightly in Davidson’s solution (BBC Biochemical, Mount Vernon, WA) for 24 controlled blood glucose levels in all strains (63–89 mg/dL; Fig. hours. After fixation, the eyes were transferred to 70% alcohol, pro- 1A). Intraocular levels were also measured to determine glu- cessed routinely for paraffin embedding, and sectioned at 4 ␮m. For cose concentration adjacent to the neural retina. Intraocular routine histopathologic evaluation the sections were stained with levels were similar to those in blood in diabetic and control hematoxylin and eosin. For immunohistochemistry, paraffin-embedded animals (Fig. 1B), suggesting that ocular humor is a major route sections were placed on charged slides and dried overnight at room of retinal glucose exposure in diabetes. Of note, SD rats temperature. After deparaffination and rehydration, slides were placed showed moderately higher levels of intraocular glucose than in citrate plus buffer (Scytek, Logan, UT) and brought to 125°C and 10 did the other two strains, a difference that reached statistical to 15 psi for 30 seconds in a Pascal pressure cooker, for antigen significance (P Ͻ 0.05). Glucose levels in the blood of diabetic retrieval, followed by washing in phosphate-buffered saline Tween animals, however, did not show consistent significant differ- (PBST) buffer (Scytek). Immunohistochemistry staining was performed ences over time between the three strains. Control BN, LE, and at room temperature with an autostainer (Dako, Carpinteria, CA). SD rats gained weight during the 3 months of observation, Tissue slides were treated for 10 minutes with 3% hydrogen peroxide however, the BN strain showed significantly less weight gain. (VWR, West Chester, PA), washed briefly with distilled water, treated In comparison, hyperglycemic rats of all strains failed to gain with a blocking agent (SuperBlock; Scytek) for 10 minutes, then re- weight over the same period (Fig. 1C). These data show that acted with mouse-anti-FGF-2 antibody (Clone bFM-2; Millipore, Bil- STZ induces hyperglycemia in blood and the eye in all three rat

Downloaded from iovs.arvojournals.org on 10/01/2021 5398 Kirwin et al. IOVS, November 2009, Vol. 50, No. 11

strains, and therefore indicate that any strain-specific molecular or functional changes in the retina are not due to large differ- ences in glucose exposure. Protein Measurements in Normal and Diabetic Rat Retinas A modified bio-assay (Luminex) was used to measure the levels of 57 proteins of interest in retinas of normal and STZ-treated BN, LE, and SD rats 1 month and 3 months after diabetes induction. Of these 57 proteins, 24 were below detection limits in normal and STZ-treated retinas, and the concentration of 22 proteins, though present at detectable levels, was not significantly changed by STZ treatment (Table 1). Hyperglycemia-Induced Retinal Inflammation in SD Rats Several markers of inflammation have been reported in the neural retina in experimental models of diabetes37,38 and in diabetic human vitreous39–41 and tissues.42–44 Although there was no evidence of a measurable inflammatory response in diabetic LE and BN rats, SD rats did show a time-dependent increase in several inflammatory proteins. The chemokine eotaxin (CCL11) was significantly increased in diabetic SD retina after 1 month to 1.6 times the level in control retina. Although the same trend could be observed at 3 months, this change was not statistically significant (P ϭ 0.052; Fig. 2A). Eotaxin was also significantly upregulated in serum after 3 months of diabetes (Table 2), as well as at 4 weeks (data not shown) in SD rats. Macrophage colony-stimulating factor (M- CSF), monocyte chemoattractant protein-1 (MCP-1; CCL-2), and MCP-3 (CCL-7) which act on monocytes and macrophages, were significantly upregulated after 3 months of diabetes in SD rats (Figs. 2B–D). Although protein levels of these cytokines appeared to be low compared with their levels in overtly

TABLE 1. Proteins Not Significantly Changed in Diabetic Retina

Detectable (but unchanged) Undetectable

Apolipoprotein A1 CD40,CD40L Calbindin EGF Endothelin-1 GCP-2 Factor VII GM-CSF GST␮ Growth Hormone IgA GST␣ ILϪ1␣,Ϫ1␤,Ϫ2,Ϫ5,Ϫ10 InterferonϪ␥ Leukemia inhibitory factor ILϪ3,Ϫ4,Ϫ7,Ϫ11,Ϫ12,Ϫ17 MIPϪ1␣,Ϫ3␤ KC/Gro␣ Myeloperoxidase Leptin Myoglobin Lymphotactin NGAL MCP-5 Stem Cell Factor MIPϪ1␤,Ϫ1␥,Ϫ2 SGOT MMP-9 Tissue Factor Oncostatin M Thrombopoietin RANTES VCAM

Proteins in the retina were not significantly affected by hypergly- cemia. Complete panel of proteins tested in all strains at 4 weeks and three months,which did not show changes or were below the detect- able level in the retina. FIGURE 1. Body weight, blood glucose, and vitreous glucose levels in Abbreviations: GST, glutathione transferase; Ig, immunoglobulin; BN, LE, and SD rats. (A) Blood glucose levels in rats of different strains IL, interleukin; MIP, macrophage inflammatory protein; NGAL, neutro- injected with STZ (solid line, filled symbols) or buffer only (broken phil gelatinase-associated lipocalin; SGOT, serum glutamic oxaloacetic line, open symbols) at multiple time points after injection. (B) Ocular transaminase; VCAM, vascular cell adhesion molecule; EGF, epidermal glucose levels at the time of death measured in the eye cup after lens growth factor; GCP, granulocyte chemotactic protein; GM-CSF, granu- removal. (C) Body weight of rats with induced diabetes and control locyte macrophage colony-stimulating factor; MMP, matrix metallopro- rats. Data are the average of results in at least three experiments with teinase; RANTES, regulated upon activation, normal T-cell expressed three rats per group (n Ͼ 9). Error bars, SD. and secreted.

Downloaded from iovs.arvojournals.org on 10/01/2021 IOVS, November 2009, Vol. 50, No. 11 Inflammation in the Retina of Diabetic Rats 5399

FIGURE 2. Retinal cytokine protein expression at 28 and 84 days of dia- betes and in age-matched control rats. The concentration of eotaxin (A) was increased in SD rats after 4 weeks of diabetes. Protein levels of M-CSF (B), MCP-1 (C), and MCP-3 (D) were increased in SD rats after 3 months of diabetes. Protein expres- sion was measured in three indepen- dent experiments. Each symbol rep- resents data from three pooled diabetic retinas of three individual diabetic or control rats. Statistical significance was determined with a Student’s t-test, and significant prob- abilities are noted.

inflammatory diseases, the increase is statistically significant inflammatory response. Clusterin and vWF also showed signif- and highly reproducible. icantly increased levels in the serum of SD rats at 3 months, Further evidence of inflammation in the retina of SD rats whereas none of the other proteins that were significantly was the upregulation of clusterin after 4 weeks and 3 months changed in the retina were altered in the serum at this time of diabetes (Fig. 3A). These results confirm published reports (Table 2). These data indicate subtle, but highly reproducible of increased clusterin expression in the diabetic SD rat,45 but and strain-dependent, inflammatory protein increases in the show the observation to be highly strain dependent. Tissue retinas of diabetic rats. inhibitor of metalloproteinase (TIMP)-1 showed a similarly moderate, but significant upregulation after 4 weeks of diabe- Changes in Growth Factor Expression in the tes and more robust changes after 3 months (Fig. 3B). Although Retinas of Diabetic Rats this protein was increased in all SD diabetic samples compared Growth factors, particularly those with angiogenic effects, are with the control, the concentration in diabetic retinas varied of interest because of their role in proliferative diabetic reti- widely between different experiments. Therefore, the Mann- nopathy.46 In BN rats, there was a small but significant increase Whitney U test was used to compare the median values be- in vascular endothelial growth factor (VEGF) protein expres- tween the diabetic and control group, showing a significant ␤ sion in the retina after 4 weeks of diabetes. However, a signif- increase. In addition, -2 microglobulin (B2M; Fig. 3C) and von icant increase was not observed after 3 months. Although there Willebrand factor (vWF; Fig. 3D) were significantly upregu- was a trend toward increased VEGF levels in LE and SD rats at lated after 3 months of diabetes, further indicating an ongoing 4 weeks, it did not reach statistical significance (P ϭ 0.115 and P ϭ 0.057 respectively; Fig. 4A). FGF-9 exhibited the same temporal pattern as VEGF, though induction of this growth TABLE 2. Serum Protein Levels factor was of a greater magnitude in BN rats. By 3 months of Diabetic Control diabetes, FGF-9 levels in the retina of diabetic rats were similar to that in control rats (Fig. 4B). In contrast, and confirming B2M (␮g/mL) 69 (Ϯ14) 65 (Ϯ11) previously published data,47 FGF-2 was significantly increased Clusterin (␮g/mL) 183 (Ϯ37)* 118 (Ϯ30) in the retina of diabetic SD rats after 3 months. This increase in Eotaxin (pg/mL) 1401 (Ϯ343)* 811 (Ϯ126) FGF-2 protein was observed only in SD rats and, although FGF-9 (ng/mL) 0.3 (Ϯ0.7) 0.4 (Ϯ0.5) Ϯ Ϯ measurable, FGF-2 levels were unchanged in BN or LE rats FGF-2 (ng/mL) 2.1 ( 0.1) 2.3 ( 0.2) (Fig. 4C). MCP-1 (pg/mL) 696 (Ϯ290) 846 (Ϯ91) MCP-3 (pg/mL) 386 (Ϯ128) 468 (Ϯ61) M-CSF (ng/mL) 0.8 (Ϯ0.1) 0.8 (Ϯ0.1) Histopathologic Changes after 3 Months TIMP-1 (ng/mL) 4.4 (Ϯ4.7) 3.3 (Ϯ3.5) of Diabetes Ϯ Ϯ VEGF (pg/mL) 238 ( 44) 211 ( 70) No gross disease was found in any of the rat strains by histo- vWF (ng/mL) 532 (Ϯ165)* 195 (Ϯ44) logic examination (Figs. 5A–F and Ref. 48), and there was no Levels of proteins (ϮSD) significantly changed in the retina of change in retinal thickness after 3 months of diabetes (data not diabetic rats. Serum was collected from SD rats after 3 months of shown). Despite an increase in inflammatory proteins in SD diabetes and protein levels determined. Results are average of three rats, no inflammatory infiltrate was detected in the retinas of separate samples each from two independent experiments (n ϭ 6). any strain (Figs. 5A–F). Staining with specific antibodies also * P Ͻ 0.05 compared with control. revealed no evidence of macrophages, CD4 T cells, CD8 T

Downloaded from iovs.arvojournals.org on 10/01/2021 5400 Kirwin et al. IOVS, November 2009, Vol. 50, No. 11

FIGURE 3. Expression of inflamma- tory proteins in the diabetic rat ret- ina. Concentrations of proteins up- regulated during inflammation were measured in the retina as in Fig. 2. In SD rats, both clusterin (A) and TIMP-1 (B) were upregulated 4 weeks and 3 months after the onset of diabetes. B2M (C) and vWF (D) were increased after 3 months of di- abetes in SD rats. Each symbol rep- resents data from three pooled dia- betic or control rats. A Student’s t-test was used to determine statisti- cal significance. *Mann-Whitney U test.

cells, or B cells in either control or diabetic retinas (data not strains in the study of diabetic retinopathy and demonstrates shown). Immunohistochemistry for FGF-2 localized the protein the highly strain-dependent nature of this animal model. to the inner nuclear layer (INL) in the retina of control and STZ-induced diabetes in rats and mice has been used exten- diabetic SD rats, and showed a dramatic increase in FGF-2 sively to study DR. In the rat model, SD rats are a commonly expression in the photoreceptor nuclear layer (PNL) after 3 used strain. However, the lack of pigmentation in these albino months of diabetes. These data suggest subtle inflammatory rats clearly interferes with the function of the visual system,50 changes in the absence of peripheral immune cells in the retina and is therefore of limited use in some studies, particularly of diabetic SD rats that coincide with FGF-2 upregulation in those examining visual function. photoreceptors. In the present study, measurement of a large number of cytokines and growth factors as well as other inflammatory Effect of Hyperglycemia on Gene Expression markers showed that most proteins were either below detect- in Photoreceptors able limits or unchanged in the eyes of diabetic rats. These Gene expression in the retina was normalized to ␤-actin and results match prior published data that failed to show a large compared in diabetic and control BN, LE, and SD rats, to scale inflammatory response in this model. In fact, in BN and LE evaluate the effect of hyperglycemia on photoreceptor func- rats, there is no evidence of cytokine upregulation through tion. After 3 months of diabetes, probe arrays (TaqMan; ABI) three months of diabetes. However, SD rats show signs of a revealed a decrease in photoreceptor-specific (Opn1mw) subtle, but reproducible inflammatory response. Four weeks mRNA in all three strains, suggesting reduced photoreceptor after the induction of diabetes, eotaxin, a basophile and eosin- function.49 (Arr3) and kinase (Grk1) ex- ophil chemoattractant, was upregulated in the diabetic retina pression are reduced in BN and SD, but not LE rats, further of SD rats compared with their nondiabetic counterparts. Al- indicating strain-dependent differences in the diabetic retina though increased eotaxin expression in the retina was tran- beyond the inflammatory response. Transducin (Gnb3) mRNA sient, significantly elevated levels were present in the serum of was increased in the retina of diabetic SD rats only (Fig. 6). The diabetic rats after 3 months of diabetes. This excludes serum as decrease of inhibitory molecules arrestin and the source of eotaxin in the retina, since the levels in this tissue mRNA contemporaneous with an upregulation of transducin are decreased at a time when serum levels hit their peak. gene interaction may indicate a compensatory mechanism in Increased protein levels of M-CSF, which act on monocytes and SD rat phototransduction. stimulate their growth, further indicate a response tailored to early immune mediators. This upregulation in SD rats occurred at the same time as increased MCP-1 and -3 protein expression, DISCUSSION cytokines that attract early inflammatory cells such as mono- In this study, we report inflammatory responses in the retina of cytes.51,52 Although MCP-3 is undetectable in vitreous or diabetic SD rats, but not LE or BN strains. Over 50 inflammatory serum of diabetic patients,53 MCP-1 has been found to be proteins and growth factors in the retina of diabetic BN, LE, increased in the vitreous of patients with diabetic retinopa- and SD rats were analyzed, and the data showed a subclinical thy41,53–55 and its level correlates with the presence of reti- inflammatory response in the retina of diabetic SD rats that nopathy.56 These inflammatory changes are subtle, and con- coincided temporally with the upregulation of FGF-2 in pho- centrations of all four cytokines are generally low, but the toreceptors as well as evidence of photoreceptor dysfunction. results are highly reproducible and clearly restricted to only It is the first direct comparison of three commonly used rat one rat strain. Despite the lack of overt inflammation, there are

Downloaded from iovs.arvojournals.org on 10/01/2021 IOVS, November 2009, Vol. 50, No. 11 Inflammation in the Retina of Diabetic Rats 5401

clusterin was overexpressed through 3 months of hyperglyce- mia and indicated early retinal damage. Of interest, though control levels of clusterin were similar between rat strains, there was no increase in either BN or LE rats. Like clusterin, TIMP-1 also showed a small but significant upregulation after 4 weeks of diabetes followed by a more robust change after 3 months in SD rats. Protein levels in the other two rat strains were low in both control and diabetic rats. Increased mRNA of the TIMP-1 ligand MMP-9 have been reported in SD rats,57 and although the proteins can be found in humans in normal interphotoreceptor matrix58 an increase correlates with vitre- ous hemorrhage in diabetic patients.59 The upregulation of B2M was another indicator of an ongoing immune response in SD rats, as was the increase in vWF after 3 months. Although some of the proteins upregulated in the retina were also in- creased in serum, these phenomena were most likely indepen- dent of each other. Only 3 of the 11 proteins that changed in the retina were also upregulated in serum and more important, eotaxin levels did not reach their peak in serum until 3 months of diabetes, a time at which retina levels had already decreased. The flow cytometry-based technique (IS 100 system; Lumi- nex) used in this study did not measure a significant increase in total VEGF in the diabetic retina of SD or LE rats at the times tested, but did show an increase in BN retina. This difference may be due to a less robust VEGF upregulation in SD than in BN rats, which has been demonstrated in retinal ischemia.8,9 Fur- thermore, vascular permeability in STZ-induced diabetes has been reported to be an early and transient phenomenon in SD rats, whereas the effect is prolonged in BN rats.10 Similar to VEGF expression, FGF-9 protein was upregulated in BN rat retinas, and this effect was transient and only observed at 4 weeks. FGF-2, on the other hand, was upregulated after 3 months in SD rats, confirming published reports.47 Immunohistochemis- try showed no inflammatory infiltrates in the retina, suggesting low-grade inflammation most likely originating in the retina. The amount of cytokines produced is evidently not high enough to induce transmigration of leukocytes into the retina. This result is in agreement with published data in both animals and patients, where increased leukostasis12,60 by neutrophils and macrophages42,60 in retinal vessels is reported without convincing evidence of extravasation of leukocytes into the retina. Although Miyamoto et al.61 report transmigration, the data presented do not lend strong support to such a claim. Immunohistochemistry further showed that FGF-2 is consti- tutively expressed in the INL, and the increase in FGF-2 was due to upregulation restricted to the PNL. Studies in other animal models have shown that FGF-2 is present in Mu¨ller cells and astrocytes,62 but is upregulated in response to injury in the retina only in photoreceptors,62,63 and this upregulation has a protective effect on these cells.64 Although there is a signifi- cant increase in apoptotic photoreceptors in diabetic SD rats, most of the photoreceptors appear unaffected.65 To compare photoreceptor viability between diabetic and control rats, genes expressed exclusively in photoreceptors were assessed. This gene expression has been shown to correlate to photore- FIGURE 4. Changes in growth factor expression in the hyperglycemic ceptor condition.66 Opn1mw mRNA was decreased in photo- rat retina. (A) A small, but highly reproducible increase in VEGF receptors of all strains, suggesting degeneration of these cells. expression was found in BN rats after 4 weeks of diabetes. (B) FGF-9 Furthermore, rhodopsin kinase and arrestin3 mRNA, which are was also increased in BN after 1 month, but not significantly changed also expressed exclusively in the photoreceptors in the ret- in any other strain or time point. (C) Protein levels of FGF-2 were 67,68 elevated in SD rats after 3 months of diabetes. Each symbol represents ina, were downregulated in BN and SD rats, but not in LE data from three pooled diabetic or control retinas. Statistical signifi- rats. Finally, Gnb3 expression increased in diabetic retina of cance was determined using Student’s t-test. SD, but not in BN or LE rats. These data suggest a negative effect of high glucose on activation of the phototransduction pathway as demonstrated by reduced opsin mRNA while rein- other indicators that hyperglycemia activates an immune re- forcing the notion of strain-dependent differences in the retina sponse in SD rats as early as 4 weeks after the induction of as seen in rhodopsin kinase, arrestin3, and Gnb3 expression. diabetes. Similar to an earlier report in diabetic SD rats,45 The reduced opsin expression may explain the reported reduc-

Downloaded from iovs.arvojournals.org on 10/01/2021 5402 Kirwin et al. IOVS, November 2009, Vol. 50, No. 11

FIGURE 5. H&E stain of the retina and localization of FGF-2. H&E stain- ing of retinas from diabetic (A, C, E) and control (B, D, F) rats after 3 months of diabetes in BN (A, B), LE (C, D), and SD (E, F) showed no inflammatory infiltrates or changes in retinal thickness in any of the sec- tions. FGF-2 immunostaining reveals an increase in this protein localized to photoreceptor nuclear layer (PNL) in diabetic SD rats (H) com- pared with control animals (G). Re- sults are representative of two inde- pendent experiments with three rats per group.

tion in photoreceptor activation with high flash intensity ERG The results of the present study clearly show that genetic in diabetic SD rats.69 However, the same study did not find an background plays a significant role in the response to hyper- effect on photoreceptor deactivation69 suggesting that the re- glycemia in the retina and is consistent with findings in other duction in rhodopsin kinase and arrestin3, although statistically studies of hyperglycemic and hypoxic stress in the retina.8–10 significant, does not affect function after 3 months of diabetes. This strain-dependent response is not due to differences in Of interest, mRNA of Gnb3 kinase which is part of the trans- glucose exposure in either blood or the area adjacent to the ducin complex is increased in SD rats. This increase may retina, as all three strains show similar levels of hyperglycemia. indicate a compensatory mechanism in damaged photorecep- A possible explanation of the lack of inflammation in LE and BN tors of this strain. rats is the presence of melanin in the two pigmented strains as It is tempting to speculate that hyperglycemia activates opposed to the lack of it in the albino SD rats. Melanin has been retinal glial cells either directly or through the binding of shown to have antioxidant properties by acting as a scavenger advanced glycation endproduct (AGE)–modified proteins to 70,71 AGE receptors on these cells. This activation may lead to for free radicals and reactive oxygen species. Nonpig- mented RPE cells show significantly more oxidative changes production of low levels of inflammatory cytokines. Although 72 their concentration is not high enough to induce infiltration of than pigmented ones and high-dose exposure to light dam- peripheral immune cells into the retina they may play a role in ages photoreceptor outer segments to a much larger extent in 73 retinal vessel leukostasis.61 The cytokines may also constitute a albino Wistar than LE rats. It is possible that hyperglycemia danger signal to photoreceptors, inducing upregulation of by itself is not enough to induce inflammatory changes in the FGF-2 and causing decreased function of photoreceptors. Al- rat eye, but that in nonpigmented rats the increase in oxidative ternatively, the ongoing inflammatory response and high glu- stress provides a secondary stimulus that results in upregula- cose levels may influence photoreceptor gene expression di- tion of inflammatory proteins in the retina and increase of rectly. FGF-2 expression in photoreceptors in SD rats.

Downloaded from iovs.arvojournals.org on 10/01/2021 IOVS, November 2009, Vol. 50, No. 11 Inflammation in the Retina of Diabetic Rats 5403

pigment epithelium-derived factor in Brown Norway and Sprague Dawley rats contributing to different susceptibilities to retinal neovascularization. Diabetes. 2002;51:1218–1225. 9. Lu K, Zhou Y, Kaufman K, Mott R, Ma J-X. Rat strain-dependent susceptibility to ischemia-induced retinopathy associated with ret- inal vascular endothelial growth factor regulation. J Mol Endocri- nol. 2007;38:423–432. 10. Zhang SX, Ma J-X, Sima J, et al. Genetic difference in susceptibility to the blood-retina barrier breakdown in diabetes and oxygen- induced retinopathy. Am J Pathol. 2005;166:313–321. 11. Chen P, Scicli GM, Guo M, et al. Role of angiotensin II in retinal leukostasis in the diabetic rat. Exp Eye Res. 2006;83:1041–1051. 12. Miyamoto K, Hiroshiba N, Tsujikawa A, Ogura Y. In vivo demon- stration of increased leukocyte entrapment in retinal microcircu- lation of diabetic rats. Invest Ophthalmol Vis Sci. 1998;39:2190– 2194. 13. Mori F, Hikichi T, Nagaoka T, Takahashi J, Kitaya N, Yoshida A. Inhibitory effect of losartan, an AT1 angiotensin II receptor antag- onist, on increased leucocyte entrapment in retinal microcircula- tion of diabetic rats. Br J Ophthalmol. 2002;86:1172–1174. 14. Muranaka K, Yanagi Y, Tamaki Y, et al. Effects of peroxisome proliferator-activated receptor gamma and its ligand on blood- retinal barrier in a streptozotocin-induced diabetic model. Invest Ophthalmol Vis Sci. 2006;47:4547–4552. 15. Poulaki V, Joussen AM, Mitsiades N, Mitsiades CS, Iliaki EF, Adamis AP. Insulin-like growth factor-I plays a pathogenetic role in dia- betic retinopathy. Am J Pathol. 2004;165:457–469. 16. Tamura H, Miyamoto K, Kiryu J, et al. Intravitreal injection of corticosteroid attenuates leukostasis and vascular leakage in ex- FIGURE 6. Gene expression patterns of proteins associated with the perimental diabetic retina. Invest Ophthalmol Vis Sci. 2005;46: phototransduction pathway. Expression of photoreceptor-specific 1440–1444. genes as well as those essential to the phototransduction pathway 17. Ichinose K, Maeshima Y, Yamamoto Y, et al. Antiangiogenic en- were measured by real-time PCR. Levels of Opsin (Opn1mw), arrestin dostatin peptide ameliorates renal alterations in the early stage of (Arr3), rhodopsin kinase (Grk1), and transducin (Gnb3) were com- a type 1 diabetic nephropathy model. Diabetes. 2005;54:2891– pared in diabetic versus control animals. Expression is denoted in 2903. relative units normalized to Actb mRNA. Data are average of results in three independent experiments with three rats per group (n ϭ 9). 18. Ishida S, Usui T, Yamashiro K, et al. VEGF164 is proinflammatory Error bars, SE. *P Ͻ 0.05, compared with the control, by Student’s in the diabetic retina. Invest Ophthalmol Vis Sci. 2003;44:2155– t-test. 2162. 19. Kinoshita N, Kakehashi A, Inoda S, et al. Effective and selective prevention of retinal leukostasis in streptozotocin-induced dia- betic rats using gliclazide. Diabetologia. 2002;V45:735–739. The findings of this study are the first direct comparison of 20. Navaratna D, McGuire PG, Menicucci G, Das A. Proteolytic degra- the retinal expression of a large number of inflammatory pro- dation of VE-cadherin alters the blood-retinal barrier in diabetes. teins and growth factors in three rat strains. It is evident that Diabetes. 2007;56:2380–2387. responses to hyperglycemia are highly strain dependent. We 21. Skondra D, Noda K, Almulki L, et al. Characterization of azurocidin demonstrate a subtle but reproducible inflammatory response as a permeability factor in the retina: Involvement in VEGF-in- in SD rats that shows some overlap with human diabetic duced and early diabetic blood-retinal barrier breakdown. Invest retinopathy. This model may be useful in identifying the mo- Ophthalmol Vis Sci. 2008;49:726–731. lecular mechanisms of subclinical inflammation and photore- 22. Hancock HA, Kraft TW. Oscillatory potential analysis and ERGs of ceptor dysfunction in the early stages of diabetic retinopathy. normal and diabetic rats. Invest Ophthalmol Vis Sci. 2004;45: 1002–1008. References 23. Kusari J, Zhou S, Padillo E, Clarke KG, Gil DW. Effect of memantine on neuroretinal function and retinal vascular changes of strepto- 1. Mohamed Q, Gillies MC, Wong TY. Management of diabetic zotocin-induced diabetic rats. Invest Ophthalmol Vis Sci. 2007;48: retinopathy: a systematic review. JAMA. 2007;298:902–916. 5152–5159. 2. Fong DS, Aiello L, Gardner TW, et al. Diabetic retinopathy. Dia- 24. Ramsey DJ, Ripps H, Qian H. An electrophysiological study of betes Care. 2003;26:99S–102S. retinal function in the diabetic female rat. Invest Ophthalmol Vis 3. Frank RN. Diabetic retinopathy. N Engl J Med. 2004;350:48–58. Sci. 2006;47:5116–5124. 4. Antonetti DA, Barber AJ, Bronson SK, et al. Diabetic retinopathy: 25. Ramsey DJ, Ripps H, Qian H. Streptozotocin-induced diabetes seeing beyond glucose-induced microvascular disease. Diabetes. modulates GABA receptor activity of rat retinal neurons. Exp Eye 2006;55:2401–2411. Res. 2007;85:413–422. 5. Gardner TW, Antonetti DA, Barber AJ, LaNoue KF, Levison SW. 26. Joussen AM, Huang S, Poulaki V, et al. In vivo retinal gene expres- Diabetic retinopathy: more than meets the eye. Surv Ophthalmol. sion in early diabetes. Invest Ophthalmol Vis Sci. 2001;42:3047– 2002;47:S253–S262. 3057. 6. Melmed RN, Benitez CJ, Holt SJ. Intermediate cells of the pancreas: 27. Shinoda K, Rejdak R, Schuettauf F, et al. Early electroretinographic III. Selective autophagy and destruction of ␤-granules in interme- features of streptozotocin-induced diabetic retinopathy. Clin Exp diate cells of the rat pancreas induced by alloxan and streptozo- Ophthalmol. 2007;35:847–854. tocin. J Cell Sci. 1973;13:297–315. 28. Agardh C-D, Agardh E, Qian Y, Hultberg B. Glutathione levels are 7. Sheetz MJ, King GL. Molecular understanding of hyperglycemia’s reduced in diabetic rat retina but are not influenced by ischemia adverse effects for diabetic complications. JAMA. 2002;288:2579– followed by recirculation. Metabolism. 1998;47:269–272. 2588. 29. Gustavsson C, Agardh CD, Hagert P, Agardh E. Inflammatory mark- 8. Gao G, Li Y, Fant J, Crosson CE, Becerra SP, Ma J-X. Difference in ers in nondiabetic and diabetic rat retinas exposed to ischemia ischemic regulation of vascular endothelial growth factor and followed by reperfusion. Retina. 2008;28:645–652.

Downloaded from iovs.arvojournals.org on 10/01/2021 5404 Kirwin et al. IOVS, November 2009, Vol. 50, No. 11

30. Oku H, Kodama T, Sakagami K, Puro DG. Diabetes-induced dis- 52. Isoda K, Folco E, Marwali MR, Ohsuzu F, Libby P. Glycated LDL ruption of gap junction pathways within the retinal microvascula- increases monocyte CC 2 expression and ture. Invest Ophthalmol Vis Sci. 2001;42:1915–1920. monocyte chemoattractant protein-1-mediated chemotaxis. Ath- 31. Heuer JG, Bailey DL, Sharma GR, et al. Cecal ligation and puncture erosclerosis. 2008;198:307–312. with total parenteral nutrition: a clinically relevant model of the 53. Abu El-Asrar AM, Struyf S, Kangave D, Geboes K, Van Damme metabolic, hormonal, and inflammatory dysfunction associated J. Chemokines in proliferative diabetic retinopathy and prolifera- with critical illness. J Surg Res. 2004;121:178–186. tive vitreoretinopathy. Eur Cytokine Netw. 2006;17:155–165. 32. Heuer JG, Sharma GR, Gerlitz B, et al. Evaluation of protein C and 54. Murugeswari P, Shukla D, Rajendran A, Kim R, Namperumalsamy P, other biomarkers as predictors of mortality in a rat cecal ligation Muthukkaruppan V. Proinflammatory cytokines and angiogenic and and puncture model of sepsis. Crit Care Med. 2004;32:1570–1578. anti-angiogenic factors in vitreous of patients with proliferative dia- 33. Kirwin SJ, Dowdell KC, Hindinger C, et al. Altered neuroantigen- betic retinopathy and Eales’ disease. Retina. 2008;28:817–824. specific cytokine secretion in a Th2 environment reduces experi- 55. Maier R, Weger M, Haller-Schober EM, et al. Multiplex bead analysis of mental autoimmune encephalomyelitis. J Neuroimmunol. 2006; vitreous and serum concentrations of inflammatory and proangio- 178:30–39. genic factors in diabetic patients. Mol Vis. 2008;14:637–643. 34. Wang JJ, Zhang SX, Mott R, et al. Salutary effect of pigment 56. Tashimo A, Mitamura Y, Nagai S, et al. Aqueous levels of macrophage epithelium-derived factor in diabetic nephropathy: evidence for migration inhibitory factor and monocyte chemotactic protein-1 in antifibrogenic activities. Diabetes. 2006;55:1678–1685. patients with diabetic retinopathy. Diabet Med. 2004;21:1292–1297. 35. Chang Y-H, Chen P-L, Tai M-C, Chen C-H, Lu D-W, Chen J-T. Hyper- 57. Giebel SJ, Menicucci G, McGuire PG, Das A. Matrix metallopro- baric oxygen therapy ameliorates the blood-retinal barrier breakdown teinases in early diabetic retinopathy and their role in alteration of in diabetic retinopathy. Clin Exp Ophthalmol. 2006;34:584–589. the blood-retinal barrier. Lab Invest. 2005;85:597–607. 36. Liao SD, Puro DG. NADϩ-induced vasotoxicity in the pericyte- 58. Plantner JJ, Smine A, Quinn TA. Matrix metalloproteinases and containing microvasculature of the rat retina: Effect of diabetes. metalloproteinase inhibitors in human interphotoreceptor matrix Invest Ophthalmol Vis Sci. 2006;47:5032–5038. and vitreous. Curr Eye Res. 1998;17:132–140. 37. Carmo A, Cunha-Vaz JG, Carvalho AP, Lopes MC. L-Arginine transport 59. Descamps FJ, Martens E, Kangave D, et al. The activated form of in retinas from streptozotocin diabetic rats: correlation with the level gelatinase B/matrix metalloproteinase-9 is associated with diabetic of IL-1␤ and NO synthase activity. Vision Res. 1999;39:3817–3823. vitreous hemorrhage. Exp Eye Res. 2006;83:401–407. 38. Kim YH, Choi MY, Kim YS, et al. Triamcinolone acetonide protects 60. Joussen AM, Murata T, Tsujikawa A, Kirchhof B, Bursell S-E, Adamis the rat retina from STZ-induced acute inflammation and early AP. Leukocyte-mediated endothelial cell injury and death in the vascular leakage. Life Sci. 2007;81:1167–1173. diabetic retina. Am J Pathol. 2001;158:147–152. 39. Elner SG, Strieter R, Bian ZM, et al. Interferon-induced protein 10 61. Miyamoto K, Khosrof S, Bursell S-E, et al. Prevention of leukostasis and vascular leakage in streptozotocin-induced diabetic retinopa- and interleukin 8. C-X-C chemokines present in proliferative dia- thy via intercellular adhesion molecule-1 inhibition. Proc Natl betic retinopathy. Arch Ophthalmol. 1998;116:1597–1601. Acad Sci USA. 1999;96:10836–10841. 40. Maier R, Weger M, Haller-Schober EM, et al. Application of multi- 62. Kostyk SK, D’Amore PA, Herman IM, Wagner JA. Optic nerve plex cytometric bead array technology for the measurement of injury alters basic fibroblast growth factor localization in the retina angiogenic factors in the vitreous. Mol Vis. 2006;12:1143–1147. and optic tract. J Neurosci. 1994;14:1441–1449. 41. Herna´ndez C, Segura RM, Fonollosa A, Carrasco E, Francisco G, 63. Xiao M, Sastry SM, Li ZY, et al. Effects of retinal laser photocoag- Simo´ R. Interleukin-8, monocyte chemoattractant protein-1 and ulation on photoreceptor basic fibroblast growth factor and sur- IL-10 in the vitreous fluid of patients with proliferative diabetic vival. Invest Ophthalmol Vis Sci. 1998;39:618–630. retinopathy. Diabet Med. 2005;22:719–722. 64. Fontaine V, Kinkl N, Sahel J, Dreyfus H, Hicks D. Survival of 42. McLeod DS, Lefer DJ, Merges C, Lutty GA. Enhanced expression of purified rat photoreceptors in vitro is stimulated directly by fibro- intracellular adhesion molecule-1 and P-selectin in the diabetic blast growth factor-2. J Neurosci. 1998;18:9662–9672. human retina and choroid. Am J Pathol. 1995;147:642–653. 65. Barber AJ, Lieth E, Khin SA, Antonetti DA, Buchanan AG, Gardner 43. Amin RH, Frank RN, Kennedy A, Eliott D, Puklin JE, Abrams GW. TW. Neural apoptosis in the retina during experimental and hu- Vascular endothelial growth factor is present in glial cells of the man diabetes: early onset and effect of insulin. J Clin Invest. retina and optic nerve of human subjects with nonproliferative 1998;102:783–791. diabetic retinopathy. Invest Ophthalmol Vis Sci. 1997;38:36–47. 66. Nir I, Agarwal N, Papermaster DS. Opsin gene expression during 44. Abu El-Asrar AM, Dralands L, Missotten L, Al-Jadaan IA, Geboes K. early and late phases of retinal degeneration in rds mice. Exp Eye Expression of apoptosis markers in the retinas of human subjects Res. 1990;51:257–267. with diabetes. Invest Ophthalmol Vis Sci. 2004;45:2760–2766. 67. Craft CM, Whitmore DH, Wiechmann AF. Cone arrestin identified 45. Kim YS, Kim YH, Cheon EW, et al. Retinal expression of clusterin in by targeting expression of a functional family (published correc- the streptozotocin-induced diabetic rat. Brain Res. 2003;976:53–59. tion appears in J Biol Chem. 1994;269(26):17756). J Biol Chem. 46. Simo R, Carrasco E, Garcia-Ramirez M, Hernandez C. Angiogenic 1994;269:4613–4619. and antiangiogenic factors in proliferative diabetic retinopathy. 68. Lyubarsky AL, Chen CK, Simon MI, Pugh EN, Jr. Mice lacking Curr Diab Rev. 2006;2:71–98. G-protein receptor kinase 1 have profoundly slowed recovery of 47. Lowe WL, Florkiewicz RZ, Yorek MA, Spanheimer RG, Albrecht cone-driven retinal responses. J Neurosci. 2000;20:2209–2217. BN. Regulation of growth factor mRNA levels in the eyes of 69. Phipps JA, Yee P, Fletcher EL, Vingrys AJ. Rod photoreceptor diabetic rats. Metabolism. 1995;44:1038–1045. dysfunction in diabetes: activation, deactivation, and dark adapta- 48. Masuzawa K, Jesmin S, Maeda S, et al. Effect of endothelin dual tion. Invest Ophthalmol Vis Sci. 2006;47:3187–3194. receptor antagonist on VEGF levels in streptozotocin-induced dia- 70. Zareba M, Szewczyk G, Sarna T, et al. Effects of photodegradation betic rat retina. Exp Biol Med. 2006;231:1090–1094. on the physical and antioxidant properties of melanosomes iso- 49. Casson RJ, Chidlow G, Wood JPM, Vidal-Sanz M, Osborne NN. The lated from retinal pigment epithelium. Photochem Photobiol. effect of retinal ganglion cell injury on light-Induced photorecep- 2006;82:1024–1029. tor degeneration. Invest Ophthalmol Vis Sci. 2004;45:685–693. 71. Seagle B-LL, Rezai KA, Gasyna EM, Kobori Y, Rezaei KA, Norris JR. 50. Douglas RM, Alam NM, Silver BD, McGill TJ, Tschetter WW, Prusky Time-resolved detection of melanin free radicals quenching reac- GT. Independent visual threshold measurements in the two eyes tive oxygen species. J Am Chem Soc. 2005;127:11220–11221. of freely moving rats and mice using a virtual-reality optokinetic 72. Wang Z, Dillon J, Gaillard ER. Antioxidant properties of melanin in system. Vis Neurosci. 2005;22:677–684. retinal pigment epithelial cells. Photochem Photobiol. 2006;82: 51. Matsushima K, Larsen CG, DuBois GC, Oppenheim JJ. Purification 474–479. and characterization of a novel monocyte chemotactic and activat- 73. Peters S, Lamah T, Kokkinou D, Bartz-Schmidt KU, Schraermeyer ing factor produced by a human myelomonocytic cell line. J Exp U. Melanin protects choroidal blood vessels against light toxicity. Med. 1989;169:1485–1490. Z Naturforsch [C]. 2006;61:427–433.

Downloaded from iovs.arvojournals.org on 10/01/2021