Macrophages accumulate during the course of corneal neovascularization, but its mechanisms and roles still remain elusive. To address these points, we herein examined corneal neovascularization after alkali injury in mice deficient in fractalkine receptor/CX3CR1, which is normally expressed by macrophages. After alkali injury, the mRNA expression of CX3CR1 was augmented along with accumulation of F4/80-positive macrophages and Gr-1-positive neutrophils in the corneas. Compared with wild-type mice, CX3CR1-deficient mice exhibited enhanced corneal neovascularization 2 wk after injury, as evidenced by enlarged CD31-positive areas. Concomitantly, the accumulation of F4/80-positive macrophages, but not Gr-1-positive neutrophils, was markedly attenuated in CX3CR1-deficient mice compared with wild-type mice. The intraocular mRNA expression of vascular endothelial growth factor (VEGF) was enhanced to similar extents in wild-type and CX3CR1-deifient mice after the injury. However, the mRNA expression of antiangiogenic factors, thrombospondin (TSP) 1, TSP-2, and a disintegrin and metalloprotease with thrombospondin (ADAMTS) 1, was enhanced to a greater extent in wild-type than CX3CR1-deificient mice. A double-color immunofluorescence analysis demonstrated that F4/80-positive cells also expressed CX3CR1 and ADAMTS-1 and that TSP-1 and ADAMTS-1 were detected in CX3CR1-positive cells. CX3CL1 enhanced TSP-1 and ADAMTS-1, but not VEGF, expression by peritoneal macrophages. Moreover, topical application of CX3CL1 inhibited corneal neovascularization at 2 wk, along with enhanced intraocular expression of TSP-1 and ADAMTS-1 but not VEGF. Thus, these observations indicate that accumulation of CX3CR1-positive macrophages intraocularly can dampen alkali-induced corneal neovascularization by producing antiangiogenic factors such as TSP-1 and ADAMTS-1 and suggest the potential therapeutic efficacy of using CX3CL1 against alkali-induced corneal neovascularization.

Corneal neovascularization (CNV)3 can arise from various causes, including corneal infections, misuse of contact lens, chemical burn, and inflammation (1, 2, 3) and frequently leads to impaired vision. Moreover, survival rates of corneal grafts were markedly reduced when placed into vascularized recipient beds or when CNV develops in the transplanted cornea (4, 5, 6, 7, 8, 9). Thus, it is necessary to develop effective measures to treat and/or prevent CNV based on the understanding of its pathogenesis at cellular and molecular levels.

Neutrophils are presumed to be the predominant leukocyte population, which infiltrate a variety of injured organs and tissues in the early stages of inflammation. Following neutrophil accumulation, mononuclear cells such as monocytes/macrophages, lymphocytes, and plasma cells infiltrate the site of tissue injury. Normal cornea does not contain any hematopoietic cells, but in the early phase of corneal injury, preceding the occurrence of CNV, a large number of neutrophils infiltrate the cornea followed by the accumulation of mononuclear cells (10, 11, 12), as observed with the injuries of other organs. Infiltrating neutrophils are presumed to contribute to the development of CNV. However, we previously proved that alkali injury-induced CNV can occur independently of granulocyte accumulation (10).

The recruitment of leukocytes to inflammatory sites is regulated by chemokines and individual chemokines attract specific leukocyte populations through processes determined by ligand specificity and by the expression patterns of the corresponding receptors (13). Mouse monocytes/macrophages express a distinct set of chemokine receptors such as CCR2 and CCR5 on their cell surface. Infiltrating monocytes/macrophages are presumed to be a rich source of angiogenic factors and can contribute to the development of neovascularization in various tissues, including cornea (14, 15, 16, 17). We observed that a potent angiogenic factor, vascular endothelial growth factor (VEGF), was consistently detected mainly in infiltrating monocyte/macrophages but not granulocytes (10). Indeed, the lack of CCR2 and/or CCR5 gene, reduced macrophage accumulation as well as CNV formation, when chemical and mechanical denudation was applied to corneal and limbal epithelium (18, 19). In contrast, several groups have provided evidence of the antiangiogenic activities of infiltrating macrophages in choroidal neovascularization (20, 21).

Mouse monocytes/macrophages also express CX3CR1 on their surface (22, 23). Moreover, a ligand for CX3CR1, CX3CL1/fractalkine, is highly expressed on vascular endothelial cells and therefore is presumed to have a role in angiogenesis (24, 25). Furthermore, the dichotomy of monocytes/macrophages has been proposed based on their expression levels of CCR2 and CX3CR1 (26, 27, 28, 29, 30). Thus, it is tempting to speculate that CX3CR1-positive monocytes/macrophages may exhibit distinct activities in CNV, compared with CCR2-positive ones. Hence, to address this assumption, we compared the molecular pathological changes in wild-type (WT) and CX3CR1-deficient mice, involving a frequently used model of ocular corneal neovascularization, namely, alkali injury-induced CNV (18, 19).

Recombinant mouse CX3CL1 (571-MF/CF) and recombinant mouse CCL2 were obtained from R&D Systems. Rat anti-mouse F4/80 mAb (clone A3-1) and rat anti-mouse CD68 mAb (clone FA-11) were obtained from Serotec and rabbit anti-mouse CD31 polyclonal Abs (pAbs) were purchased from Abcam (ab28364). Rat anti-mouse-Ly-6G mAb (clone 1A8, catalog no. 551459) and rat anti-mouse pan-NK cell mAb (clone DX5) were obtained from BD Pharmingen. Goat anti-ADAMTS-1 (a disintegrin and metalloprotease with thrombospondin 1; L-16, sc-31080), goat anti-thrombospondin (TSP) 1 (N-20), and goat anti-CX3CL1 pAbs (M-18) were obtained from Santa Cruz Biotechnology and rabbit anti-ADAMTS-1 pAbs (ALX-210-555) were from Alexis Biochemicals. Alexa Fluor 488 donkey anti-goat IgG (H + L), donkey anti-rat IgG (H + L) as well as Alexa Fluor 594 donkey anti-rabbit IgG (H + L) were purchased from Invitrogen Life Technologies. Rabbit anti-CX3CR1 pAbs were provided by Dr. T. Imai (Kan Research Institute, Kyoto, Japan).

CX3CR1-deficient mice were prepared as described previously (25) and were a gift from Drs. P. M. Murphy and J.-L. Gao (National Institute of Allergy and Infections Diseases, National Institutes of Health, Bethesda, MD). The mice were backcrossed to BALB/c mice for more than eight generations. Pathogen-free BALB/c mice were obtained from Clea Japan and were designated as WT mice. Age- and sex-matched CX3CR1-deficient and WT mice were kept under specific pathogen-free conditions in groups of five and fed regular laboratory chow and water ad libitum. A 12-h day and night cycle was maintained during the whole course of the study. All animal experiments were performed at the Institute for Experimental Animals (Kanazawa University Advanced Science Research Center, Kanazawa, Japan) in accordance with the Association for Research in Vision and Ophthalmology Statement for the Use of Animals in Ophthalmic and Vision Research and complied with the standards set out in the Guidelines for the Care and Use of Laboratory Animals of Kanazawa University and were approved by the Committee on Animal Experimentation of Kanazawa University.

Mice were anesthetized with an i.p. injection of 1.8% (v/v) Avertin at a dose of 0.15 ml/10 g body weight. A 2-mm disc of filter paper saturated with 1 N NaOH was placed onto the right cornea of each mouse for 40 s, followed by rinsing extensively with 15 ml of PBS. The corneal epithelia were removed using a corneal knife in a rotary motion parallel to the limbus by gently scraping over the corneal surface without injuring the underlying corneal stroma (10). Erythromycin ophthalmic ointment was instilled immediately following epithelial denudation. At the indicated time intervals (days 0, 2, 4, and 7), mice were killed and the corneas were removed from both eyes. These corneas were placed immediately into RNALate (Qiagen) and kept at −86°C until total RNA extraction. In another series of experiments, mice were killed at the indicated time points (days 0, 2, 4, 14, and 28) after alkali treatment and both eyes were entirely removed from each animal. These eyes were fixed in 10% neutral formalin buffer for histological analysis. The left eye of each mouse was used as an untreated control. In some experiments, recombinant CX3CL1 protein was dissolved in 0.2% sodium hyaluronate (Sigma-Aldrich) at 2.5 μg/ml. Five microliters of CX3CL1 preparation or vehicle was applied topically to the alkali-treated eye twice a day for 7 days. Each experiment was repeated at least three times.

Eyes were examined under a Lecia MZ16 surgical microsystem 14 days after alkali injury. In brief, under anesthesia, photographs of the corneas were obtained using a Leica DFC350FX digital camera that was linked to an operating microscope. Microscopic assessment was done by two independent observers without prior knowledge of the experimental procedures.

The paraffin-embedded tissues were cut into 5-μm- thick slices, mounted on poly-l-lysine-coated slides, and subjected to H&E staining. For immunohistochemical staining, the deparaffinized sections or fixed cryosections (8-μm thick) were used. Endogenous peroxidases were quenched in 0.3% (v/v) hydrogen peroxide for 10 min. For Ag retrieval, sections were treated with 0.1% trypsin in 0.1% CaCl2 for 20 min at 37°C or immersed in 10 mM sodium citrate buffer (pH 6.0) and heated for 20 min in a microwave oven. After washing with PBS, slides were incubated with blocking reagent for 20 min. For the detection of macrophages, the sections were incubated overnight at 4°C with rat anti-mouse F4/80 Ab (1 μg/ml) or rat anti-mouse CD68 Ab (1 μg/ml). Tissue sections were then incubated with biotin-conjugated anti-rat Ig Ab as secondary Abs. To identify neutrophils and NK cells, the sections were incubated overnight at 4°C with purified rat anti-mouse-Ly-6G (2.5 μg/ml) and purified rat anti-mouse pan-NK cell mAbs (2.5 μg/ml), respectively. The sections were further incubated with biotin-conjugated rabbit anti-rat Ig Ab or biotin-conjugated rabbit anti-rat IgM Ab as the secondary Abs. Other sets of sections were incubated overnight at 4°C with rabbit anti-CX3CR1 and goat anti-CX3CL1 pAbs to detect CX3CR1 and CX3CL1 expression, respectively. These slides were incubated with biotin-conjugated anti-rabbit or anti-goat Ig Ab as the secondary Abs. The immune complexes were detected by using an ABC kit and a DAB Substrate Kit from Vector Laboratories according to the manufacturer’s instructions. Slides were then counterstained with hematoxylin and mounted. The numbers of positive cells were counted on five randomly chosen fields of corneal sections in each animal at 200-fold magnification, by an examiner without any prior knowledge of the experimental procedures. The numbers of positive cells per mm2 were calculated.

The deparaffinized sections (5-μm thick) or fixed cryosections (8-μm thick) were stained using anti-CD31 pAbs and the numbers and sizes of the CNV were determined as described previously (10, 31, 32) by an examiner with no knowledge of the experimental procedures. Briefly, images were captured with a digital camera and imported into Adobe Photoshop (version 7.0). Then, the number of neovascular tubes per mm2 and the proportions of CNV in the hot spots were determined using NIH Image analysis software version 1.62 (National Institutes of Health, Bethesda, MD). Most sections were from the central region of the cornea. The numbers and areas of CNV were evaluated on at least two sections from each eye.

Total RNAs were extracted from the corneas or cultured peritoneal macrophages with the RNeasy Mini Kit (Qiagen) according to the manufacturer’s instructions. The RNA preparations were further treated with RNase-free DNase I (Invitrogen Life Technologies) to remove residual genomic DNA. Two micrograms of total RNA was reverse-transcribed at 42°C for 1 h in 20 μl of reaction mixture containing mouse Moloney leukemia virus reverse transcriptase and hexanucleotide random primers (Qiagen). Real-time PCR was performed on an Applied Biosystems StepOne Real-Time PCR System using the comparative threshold cycle (CT) quantification method. TaqMan Gene Expression Assays (Applied Biosystems) containing specific primers (accession numbers: CX3CL1, Mm00436454_ml; CX3CR1, Mm02620111_s1; TSP-1, Mm00449032_gl; TSP-2, Mm00449036_m1; ADAMTS-1, Mm00477355_m1; VEGF, Mm00437304_m1; TGF-β1, Mm03024053_m1; GAPDH, Mm99999915_ g1), and TaqMan MGB probe (FAM dye labeled)/TaqMan Fast Universal PCR Master Mix were used with 10 ng of cDNA to detect and quantify the expression levels of CX3CL1, CX3CR1, TSP-1, TSP-2, ADAMTS-1, VEGF, and TGF-β1. Reactions were performed for 20 s at 95°C, then with 40 cycles of 1 s at 95°C and 20 s at 60°C. GAPDH was amplified as an internal control. CT values of GAPDH were subtracted from CT values of the target genes (ΔCT). ΔCT values of WT mice were compared with ΔCT values of CX3CR1-deficient mice.

Mononuclear cells were isolated from corneas according to the procedure described previously by Ohshima et al. (33), with some modifications. At 2 days after the alkali injury, corneas were removed, teased away with scissors, and incubated at 37°C for 30 min with constant shaking in the presence of 0.5 mg/ml collagenase type D (Roche Diagnostics). Cell suspensions were then passed over a nylon filter with a 100-μm pore size. The resultant cells were further layered on Histopaque-1083 (Sigma-Aldrich) and were centrifuged to obtain mononuclear cells. Isolated cells were stained with FITC-conjugated rat anti-mouse F4/80 mAb and rabbit anti-CX3CR1 pAbs followed by staining with Alexa Fluor 647-conjugated goat anti-rabbit IgG. Fluorescence intensities were determined with the help of a FACSCalibur (BD Biosciences), along with staining of the samples with nonimmunized rabbit IgG (Sigma-Aldrich) as a negative control.

Sections were deparaffinized and treated with 0.1% trypsin for 15 min at 37°C for Ag retrieval. Thereafter, the sections were incubated with the combinations of rat anti-mouse F4/80 and rabbit anti-mouse CX3CR1, rat anti-F4/80 and rabbit anti-ADAMTS-1, goat anti-ADAMTS-1 and rabbit anti-mouse CX3CR1, or goat anti-TSP-1 and rabbit anti-mouse CX3CR1 Abs overnight at 4°C. After being rinsed with PBS, for a double-immunofluorescence analysis using anti-F4/80, the sections were then incubated with the combination of Alexa Fluor 488 donkey anti-rat IgG and Alexa Fluor 594 donkey anti-rabbit IgG (1/100) for 40 min at room temperature in the dark. For a double-color immunofluorescence analysis of CX3CR1 and antiangiogenic factors, the sections were further incubated with the combination of Alexa Fluor 488 donkey anti-goat IgG and Alexa Fluor 594 donkey anti-rabbit IgG (1/100) for 40 min at room temperature in the dark. Finally, the sections were washed with PBS and immunofluorescence was visualized in a dual-channel mode on an Olympus fluorescence microscope. Images were processed using Adobe Photoshop software version 7.0.

Specific pathogen-free 8- to 10-wk-old male WT or CX3CR1-deficient mice were injected i.p. with 2 ml of sterile 3% thioglycolate medium (Sigma-Aldrich), and i.p. macrophages were harvested 3 days later as described previously (34). The resultant cell preparation consists of >95% macrophages as verified by using flow cytometric analysis on the cell preparation immunostained with anti-F4/80 Ab. The cells were suspended in antibiotic-free RPMI 1640 medium containing 10% FBS, and incubated in a humidified incubator at 37°C in 5% CO2 in 24-well cell culture plates. Two hours later, nonadherent cells were removed and the medium was replaced. The cells were then stimulated with the indicated concentrations of murine CX3CL1 or CCL2 for 12 h. Total RNAs were extracted from the cultured cells and subjected to quantitative RT-PCR as described above. For an immunocytochemical analysis of ADAMTS-1 expression, murine macrophages were seeded onto the wells of a Lab-Tec chamber slide with eight wells (Nalge Nunc) at 5 × 104 cells/well. After adhesion, the cells were stimulated with the indicated concentration of murine CX3CL1 for 24 h in a 37°C incubator with 5% CO2 and then subjected to immunocytochemical study as previous described (35).

The means and SEM were calculated on all parameters determined in the study. Data were analyzed statistically using one-way ANOVA or two-tailed Student’s t test. A value of p < 0.05 was accepted as statistically significant.

Normal corneas do not contain any leukocytes or vascular structures. In line with our previous observations (10), alkali injury markedly increased the numbers of F4/80-positive macrophages and Gr-1-positive neutrophils in the cornea, reaching maximal levels at 4 days after the injury and decreasing thereafter (Fig. 1). Most of the infiltrated mononuclear cells observed in H&E-stained tissue samples were identified as macrophages by morphological criteria (data not shown). Only a few NK cells were detected after the injury (day 2, undetectable; day 4, 8.8 ± 3.0/mm2; day 7, 4.3 ± 2.4/mm2). Because CX3CR1 is expressed by a proportion of macrophages, we examined the expression of CX3CR1 and its ligand CX3CL1 in corneas after alkali-induced injury with the use of quantitative RT-PCR. CX3CR1 mRNA was barely detected in untreated eyes, but its expression was enhanced in eyes 2 days after injury and further augmented 7 days after the injury (Fig. 2,A). The mRNA of a specific CX3CR1 ligand, CX3CL1, was constitutively expressed in the eyes before and after the injury. CX3CL1 proteins were consistently expressed by corneal epithelial and corneal endothelium, the cell that is localized in the inner layer of the cornea and is distinct from vascular endothelium and, to a lesser degree, by corneal stroma of untreated WT mice. At 2–4 days after the injury, the time when a large number of macrophages infiltrated intracorneally, CX3CL1 was additionally detected in the infiltrating cells (Fig. 2,B, lower panel). CX3CR1-positive cells were not detected in untreated WT mice but appeared in the cornea 2 or 4 days after the injury (Fig. 2, B, upper panel, and C). Moreover, a majority of mononuclear cells, present in cornea 2 days after the injury, expressed F4/80 (data not shown) and a substantial proportion of these F4/80-positive cells expressed CX3CR1 (Fig. 2 D). These observations would indicate that alkali injury induced intracorneal accumulation of F4/80-positive cells and some of them expressed CX3CR1.

FIGURE 1.

Changes with time in each population of intracorneally accumulating inflammatory cells after alkali injury. The harvested corneas were analyzed by immunohistochemical analysis at the indicated time points after alkali injury. The numbers of corneal infiltrating inflammatory cells per mm2 were calculated as described in Materials and Methods. All values represent means ± SEM of three to five independent measurements.

FIGURE 1.

Changes with time in each population of intracorneally accumulating inflammatory cells after alkali injury. The harvested corneas were analyzed by immunohistochemical analysis at the indicated time points after alkali injury. The numbers of corneal infiltrating inflammatory cells per mm2 were calculated as described in Materials and Methods. All values represent means ± SEM of three to five independent measurements.

Close modal
FIGURE 2.

A, Quantitative RT-PCR to assess mRNA expression of CX3CR1 and its ligand CX3CL1 using the obtained total RNAs from corneas at the indicated time points as described in Materials and Methods. Representative results from three independent experiments are shown here. B, CX3CR1 and CX3CL1 protein expression in injured corneas of WT mice. Corneal tissues were obtained 0, 2, 4, and 7 days after the injury from WT mice. Tissues were stained with anti-CX3CR1 Abs (upper panels) and anti-CX3CL1 Abs (lower panels). Representative results from five individual animals are shown here. Original magnification, ×400. Scale bar, 50 μm. C, The numbers of infiltrated CX3CR1-positive cells were determined as described in Materials and Methods and mean and SEM are shown here (n = 5). D, The CX3CR1 expression on the accumulated F4/80-positive macrophages 4 days after the injury was determined by a flow cytometric analysis. Purified mononuclear cells were stained with FITC-conjugated rat anti-mouse F4/80 Abs and rabbit anti-CX3CR1 Abs (open heavy-lined histogram) or nonimmunized rabbit IgG (filled histogram) as a negative control followed by staining with Alexa Fluor 647-conjugated goat anti-rabbit IgG. Most mononuclear cells were positive for F4/80. A representative result from three independent experiments is shown.

FIGURE 2.

A, Quantitative RT-PCR to assess mRNA expression of CX3CR1 and its ligand CX3CL1 using the obtained total RNAs from corneas at the indicated time points as described in Materials and Methods. Representative results from three independent experiments are shown here. B, CX3CR1 and CX3CL1 protein expression in injured corneas of WT mice. Corneal tissues were obtained 0, 2, 4, and 7 days after the injury from WT mice. Tissues were stained with anti-CX3CR1 Abs (upper panels) and anti-CX3CL1 Abs (lower panels). Representative results from five individual animals are shown here. Original magnification, ×400. Scale bar, 50 μm. C, The numbers of infiltrated CX3CR1-positive cells were determined as described in Materials and Methods and mean and SEM are shown here (n = 5). D, The CX3CR1 expression on the accumulated F4/80-positive macrophages 4 days after the injury was determined by a flow cytometric analysis. Purified mononuclear cells were stained with FITC-conjugated rat anti-mouse F4/80 Abs and rabbit anti-CX3CR1 Abs (open heavy-lined histogram) or nonimmunized rabbit IgG (filled histogram) as a negative control followed by staining with Alexa Fluor 647-conjugated goat anti-rabbit IgG. Most mononuclear cells were positive for F4/80. A representative result from three independent experiments is shown.

Close modal

Given the fact that CX3CR1 is expressed on macrophages (22, 23), we examined the effects of CX3CR1 deficiency on leukocyte accumulation in injured corneas. Ly-6G-positive neutrophils accumulated to a similar extent in the corneas of WT and CX3CR1-deficient mice after the injury (data not shown). On the contrary, the number of F4/80-positive cells was markedly depressed in CX3CR1-deficient mice compared with WT mice (Fig. 3). When another macrophage marker, CD68, was used to identify macrophages, similar results were obtained (Fig. 3). Thus, the CX3CL1-CX3CR1 interactions may regulate F4/80- and CD68-positive macrophage, but not neutrophil accumulation in damaged corneas.

FIGURE 3.

Macrophage accumulation in injured corneas of WT and CX3CR1-deficient mice. A, Corneal tissues from WT mice (left panels) or CX3CR1-deficient mice (right panels) obtained 0 (upper panels) or 4 days (middle and lower panels) after the injury were stained with rat anti-F4/80 (upper and middle panels) or rat anti-CD68 (lower panels) mAbs. Original magnification, ×400. Scale bar, 50 μm. B, The numbers of accumulated F4/80-positive or CD68-positive macrophages were determined as described in Materials and Methods and the mean and SEM are shown here (n = 5). *, p < 0.05; **, p < 0.01, WT vs CX3CR1-deficient mice.

FIGURE 3.

Macrophage accumulation in injured corneas of WT and CX3CR1-deficient mice. A, Corneal tissues from WT mice (left panels) or CX3CR1-deficient mice (right panels) obtained 0 (upper panels) or 4 days (middle and lower panels) after the injury were stained with rat anti-F4/80 (upper and middle panels) or rat anti-CD68 (lower panels) mAbs. Original magnification, ×400. Scale bar, 50 μm. B, The numbers of accumulated F4/80-positive or CD68-positive macrophages were determined as described in Materials and Methods and the mean and SEM are shown here (n = 5). *, p < 0.05; **, p < 0.01, WT vs CX3CR1-deficient mice.

Close modal

We next explored CNV in CX3CR1-deficient and WT mice. Until 7 days after the injury, CNV was not detected microscopically (data not shown). By contrast, CNV was macroscopically evident in WT mice 2 wk after the injury, as we previously reported (10). At this time point, macroscopic CNV was more enhanced in CX3CR1-deficient mice than in WT mice (Fig. 4,A). An immunohistochemical analysis using anti-CD31 Ab further demonstrated that vascular areas were increased to a larger extent in CX3CR1-deficient mice than in WT mice (Fig. 4, B and C). Moreover, we also observed that macroscopic CNV persisted in CX3CR1-deficient mice 4 wk after the injury, when CNV was resolved in WT mice (Fig. 4, B and C). These observations indicate that CX3CR1 deficiency enhanced the alkali-induced CNV, along with reduced macrophage accumulation.

FIGURE 4.

Alkali injury-induced CNV. A, Macroscopic appearance of WT (upper panels) and CX3CR1-deficient mouse eyes (lower panels) 2 wk after alkali injury. B, Corneal tissues were obtained at 2 and 4 wk after the injury from WT mice (left panels) or CX3CR1-deficient mice (right panels). Tissues were stained with H&E (upper panels) or immunostained with anti-CD31 Abs (middle and lower panels) and representative results from five individual animals are shown. Original magnification, ×400. Scale bar, 50 μm. C, CNV numbers per mm2 in hot spots (upper panel) or percent CNV areas in hot spots (lower panel) were determined on corneas obtained from WT or knockout (KO) mice 2 and 4 wk after the injury. Each value represents mean and SEM (n = 5 animals). *, p < 0.05; **, p < 0.01, WT vs CX3CR1-deficient mice.

FIGURE 4.

Alkali injury-induced CNV. A, Macroscopic appearance of WT (upper panels) and CX3CR1-deficient mouse eyes (lower panels) 2 wk after alkali injury. B, Corneal tissues were obtained at 2 and 4 wk after the injury from WT mice (left panels) or CX3CR1-deficient mice (right panels). Tissues were stained with H&E (upper panels) or immunostained with anti-CD31 Abs (middle and lower panels) and representative results from five individual animals are shown. Original magnification, ×400. Scale bar, 50 μm. C, CNV numbers per mm2 in hot spots (upper panel) or percent CNV areas in hot spots (lower panel) were determined on corneas obtained from WT or knockout (KO) mice 2 and 4 wk after the injury. Each value represents mean and SEM (n = 5 animals). *, p < 0.05; **, p < 0.01, WT vs CX3CR1-deficient mice.

Close modal

Monocytes/macrophages can be a rich source of both angiogenic and antiangiogenic factors (14, 16, 17, 20, 21, 36, 37, 38), and the balance between these two distinct sets of factors can determine the outcome of angiogenesis processes in various situations. Hence, we examined the mRNA expression of angiogenic and antiangiogenic factors in corneas after injury. The mRNA expression of two potent angiogenic factors, VEGF and TGF-β, was marginally augmented and to similar extents in WT and CX3CR1-deficient mice after the injury (Fig. 5, D and E). The intraocular mRNA expression of the antiangiogenic factors TSP-1 and ADAMTS-1 and, to a lesser extent, TSP-2, was enhanced in WT mice after the injury, but their enhanced expression was depressed in CX3CR1-deficient mice (Fig. 5, A–C). These observations imply that CX3CR1 deficiency reduced antiangiogenic factor expression and favored angiogenesis.

FIGURE 5.

Quantitative RT-PCR analysis of proangiogenic and antiangiogenic gene expression in the injured corneas of WT and CX3CR1-deficient mice. Representative results from three independent experiments are shown. The mRNAs of ADAMTS-1 (A), TSP-1 (B), TSP-2 (C), VEGF (D), and TGF-β1 (E) to GAPDH of WT (□) and CX3CR1-deficient (▪) mice at the indicated time intervals after the alkali injury were determined by quantitative RT-PCR as described in Materials and Methods. All values represent means and SEM (n = 3–5 animals). *, p < 0.05; ** p < 0.01, WT vs CX3CR1-deficient mice.

FIGURE 5.

Quantitative RT-PCR analysis of proangiogenic and antiangiogenic gene expression in the injured corneas of WT and CX3CR1-deficient mice. Representative results from three independent experiments are shown. The mRNAs of ADAMTS-1 (A), TSP-1 (B), TSP-2 (C), VEGF (D), and TGF-β1 (E) to GAPDH of WT (□) and CX3CR1-deficient (▪) mice at the indicated time intervals after the alkali injury were determined by quantitative RT-PCR as described in Materials and Methods. All values represent means and SEM (n = 3–5 animals). *, p < 0.05; ** p < 0.01, WT vs CX3CR1-deficient mice.

Close modal

In line with the previous observation that CX3CR1 is expressed on macrophages (22, 23), we observed that a substantial proportion of F4/80-positive macrophages expressed CX3CR1 (Figs. 2,D and 6,A). ADAMTS-1 protein was detected in F4/80-positive macrophages (Fig. 6,B). Moreover, CXC3CR1-positive cells, present in injured cornea, expressed antiangiogenic factors such as ADAMTS-1 (Fig. 6,C) and TSP-1 (Fig. 6 D). These observations imply that CX3CR1-positive macrophages counteracted alkali-induced CNV by expressing antiangiogenic factors such as TSP-1 and ADAMTS-1.

FIGURE 6.

A double-color immunofluorescence analysis of injured corneas. Corneas were obtained from WT mice 2 days after the injury. The samples were immunostained with a combination of anti-F4/80 and anti-CX3CR1 (A), anti-F4/80 and anti-ADAMTS-1 (B), anti-CX3CR1 and anti-ADAMTS-1 (C), or anti-CX3CR1 and anti-TSP-1 (D) Abs as described in Materials and Methods and observed under a fluorescence microscopy. Original magnification, ×400. Signals were digitally merged in the right panels. Arrows, The double positively stained cells. Representative results from three independent experiments are shown.

FIGURE 6.

A double-color immunofluorescence analysis of injured corneas. Corneas were obtained from WT mice 2 days after the injury. The samples were immunostained with a combination of anti-F4/80 and anti-CX3CR1 (A), anti-F4/80 and anti-ADAMTS-1 (B), anti-CX3CR1 and anti-ADAMTS-1 (C), or anti-CX3CR1 and anti-TSP-1 (D) Abs as described in Materials and Methods and observed under a fluorescence microscopy. Original magnification, ×400. Signals were digitally merged in the right panels. Arrows, The double positively stained cells. Representative results from three independent experiments are shown.

Close modal

Because the CX3CR1-mediated signal can enhance the functions of macrophages, we next examined the effects of exogenous CX3CL1 on antiangiogenic factor expression by murine peritoneal macrophages. CX3CL1 enhanced TSP-1 and ADAMTS-1, but not VEGF, mRNA expression by peritoneal macrophages (Fig. 7,A). CX3CL1-stimulated macrophages consistently exhibited increased ADAMTS-1 protein expression compared with untreated ones (Fig. 7, D and E). By contrast, another macrophage-tropic chemokine, CCL2, enhanced VEGF, but not TSP-1 and ADAMTS-1, mRNA expression by peritoneal macrophages obtained from WT or CX3CR1-deficient mice (Fig. 7 B). Thus, CX3CL1 can induce CX3CR1-positive macrophages to express antiangiogenic factors such as TSP-1 and ADAMTS-1, while another macrophage-tropic chemokine, CCL2, can induce CX3CR1-negative macrophages to express VEGF.

FIGURE 7.

Comparison of the levels of VEGF, TSP-1, and VEGF expression by murine peritoneal macrophages stimulated with CX3CL1 or CCL2. A, Peritoneal macrophages from WT mice were incubated with the indicated concentrations of CX3CL1 for 12 h. Quantitative RT-PCR was performed on total RNAs extracted from the macrophages as described in Materials and Methods. The levels of VEGF, ADAMTS-1, and TSP-1 mRNA were determined and normalized to GAPDH mRNA levels. Each value represents the mean and SEM (n = 3). *, p < 0.05; **, p < 0.01, compared with untreated. B, Peritoneal macrophages from WT or CX3CR1-deficient mice were incubated with CCL2 (10 ng/ml) for 12 h. Quantitative RT-PCR was performed on total RNAs extracted from the macrophages as described in Materials and Methods. The levels of VEGF, ADAMTS-1, and TSP-1 mRNA were determined and normalized to GAPDH mRNA levels. Each value represents the mean and SEM (n = 3). **, p < 0.01, compared with untreated. C–E, Peritoneal macrophages from WT mice were stimulated in the absence (D) or presence of CX3CL1 (10 ng/ml; C and E) for 24 h. The cells were then processed for immunocytochemical analysis using anti-ADAMTS-1 Abs (D and E) or control Abs (C) as described in Materials and Methods. The representative results from three independent experiments are shown. Original magnification, ×400. Scale bar, 50 μm.

FIGURE 7.

Comparison of the levels of VEGF, TSP-1, and VEGF expression by murine peritoneal macrophages stimulated with CX3CL1 or CCL2. A, Peritoneal macrophages from WT mice were incubated with the indicated concentrations of CX3CL1 for 12 h. Quantitative RT-PCR was performed on total RNAs extracted from the macrophages as described in Materials and Methods. The levels of VEGF, ADAMTS-1, and TSP-1 mRNA were determined and normalized to GAPDH mRNA levels. Each value represents the mean and SEM (n = 3). *, p < 0.05; **, p < 0.01, compared with untreated. B, Peritoneal macrophages from WT or CX3CR1-deficient mice were incubated with CCL2 (10 ng/ml) for 12 h. Quantitative RT-PCR was performed on total RNAs extracted from the macrophages as described in Materials and Methods. The levels of VEGF, ADAMTS-1, and TSP-1 mRNA were determined and normalized to GAPDH mRNA levels. Each value represents the mean and SEM (n = 3). **, p < 0.01, compared with untreated. C–E, Peritoneal macrophages from WT mice were stimulated in the absence (D) or presence of CX3CL1 (10 ng/ml; C and E) for 24 h. The cells were then processed for immunocytochemical analysis using anti-ADAMTS-1 Abs (D and E) or control Abs (C) as described in Materials and Methods. The representative results from three independent experiments are shown. Original magnification, ×400. Scale bar, 50 μm.

Close modal

Finally, we examined the effects of topically applied CX3CL1 on alkali-induced CNV. Topical administration of CX3L1 alone caused little accumulation of F4/80-positive macrophages in corneas (Fig. 8, A and B; day 2, undetectable; day 4, 16.8 ± 8.3). By contrast, topical administration of CX3CL1 augmented alkali injury-induced increases in the numbers of intracorneal macrophages. Similarly, the administration of CX3CL1 after alkali injury enhanced mRNA expression of antiangiogenic factors, TSP-1 and ADAMTS-1, but not VEGF, compared with vehicle treatment (Fig. 8,C), whereas CX3CL1 alone failed to increase the mRNA expression of these genes (data not shown). Concomitantly, at 2 wk after the injury, the alkali-induced CNV was significantly attenuated by treatment with CX3CL1, but not with vehicle (Fig. 8, D–I). These results further suggest that CX3CL1 can be effective for prevention and/or treatment for alkali-induced CNV.

FIGURE 8.

The effects of topically applied CX3CL1 on alkali-induced CNV. A and B, Corneal tissues were removed at 2 or 4 days after the injury from WT mice treated with CX3CL1 alone, alkali injury alone, or a combination of CX3CL1 and alkali injury and were stained with rat anti-F4/80 mAb. Representative results from five independent experiments are shown in A. Original magnification, ×400. Scale bar, 50 μm. The numbers of F4/80-positive macrophages were determined as described in Materials and Methods and the mean and SEM are shown in B (n = 5). **, p < 0.01, the combined treatment with CX3CL1 and alkali injury vs alkali injury. C, At the indicated time intervals after the injury, total RNAs were extracted from the cornea and processed for quantitative RT-PCR for VEGF, ADAMTS-1, and TSP-1 levels of vehicle-treated (□) and CX3CL1-treated mice (▪). Means and SEM are shown (n = 3 animals). *, p < 0.05, CX3CL1-treated vs untreated groups. D and E, Representative macroscopic appearance of vehicle-treated (D) and CX3CL1 (2.5 μg/ml) topically applied mice (E) at 2 wk after alkali injury. F and G, Cryosections from corneal tissues 2 wk after the injury from vehicle-treated (F) and CX3CL1 topically applied (G) mice were immunostained with anti-CD31 Abs and representative results from five individual mice in each group are shown. Original magnification in F and G, ×200 with a scale bar of 100 μm. H and I, The CNV numbers per mm2 in hot spots (H) or percent CNV areas in hot spots (I) were determined as described in Materials and Methods. Each value represents the mean and SEM (n = 5 animals). *, p < 0.05; **, p < 0.01, CX3CL1-treated vs untreated groups.

FIGURE 8.

The effects of topically applied CX3CL1 on alkali-induced CNV. A and B, Corneal tissues were removed at 2 or 4 days after the injury from WT mice treated with CX3CL1 alone, alkali injury alone, or a combination of CX3CL1 and alkali injury and were stained with rat anti-F4/80 mAb. Representative results from five independent experiments are shown in A. Original magnification, ×400. Scale bar, 50 μm. The numbers of F4/80-positive macrophages were determined as described in Materials and Methods and the mean and SEM are shown in B (n = 5). **, p < 0.01, the combined treatment with CX3CL1 and alkali injury vs alkali injury. C, At the indicated time intervals after the injury, total RNAs were extracted from the cornea and processed for quantitative RT-PCR for VEGF, ADAMTS-1, and TSP-1 levels of vehicle-treated (□) and CX3CL1-treated mice (▪). Means and SEM are shown (n = 3 animals). *, p < 0.05, CX3CL1-treated vs untreated groups. D and E, Representative macroscopic appearance of vehicle-treated (D) and CX3CL1 (2.5 μg/ml) topically applied mice (E) at 2 wk after alkali injury. F and G, Cryosections from corneal tissues 2 wk after the injury from vehicle-treated (F) and CX3CL1 topically applied (G) mice were immunostained with anti-CD31 Abs and representative results from five individual mice in each group are shown. Original magnification in F and G, ×200 with a scale bar of 100 μm. H and I, The CNV numbers per mm2 in hot spots (H) or percent CNV areas in hot spots (I) were determined as described in Materials and Methods. Each value represents the mean and SEM (n = 5 animals). *, p < 0.05; **, p < 0.01, CX3CL1-treated vs untreated groups.

Close modal

We previously proved that alkali-induced CNV can develop independently of neutrophil accumulation (10) in contrast to several observations, which indicate the crucial involvement of neutrophils in various types of neovascularization (11, 12, 39, 40, 41, 42, 43, 44). We observed that VEGF proteins were detected mainly in the accumulated mononuclear cells in this alkali-induced CNV (10). This may mirror the assumption that macrophages are a rich source of various angiogenic factors. Macrophages utilize various chemokine receptors such as CCR2 or CCR5 for their trafficking. Accordingly, CCR2- or CCR5-deficient mice exhibited reductions in macrophage accumulation and CNV after corneal injury (18, 19). Moreover, several lines of evidence would indicate that monocytes/macrophages can be divided into CCR2highCX3CR1low and CCR2lowCX3CR1high subpopulations (26, 27, 28, 29, 30). Indeed, a proportion of intracorneally accumulated macrophages expressed CX3CR1 in the absence of enhanced mRNA expression of its single ligand, CX3CL1, in the injured eyes. CX3CL1 is a membrane-bound chemokine and can be recycled between plasma membrane and a juxtanuclear compartment (45). Thus, it is tempting to speculate that alkali injury could increase the trafficking of CX3CL1 from a juxtanuclear compartment to the plasma membrane and eventually increase the amount of biologically active CX3CL1, which is localized in the plasma membrane. This, in turn, can induce intracorneal accumulation of CX3CR1-expressing macrophages.

These observations prompted us to investigate the roles of the CX3CL1-CX3CR1 interactions in alkali injury-induced CNV by comparing the molecular pathological changes between CX3CR1-deficient and WT mice. We observed that macrophage accumulation was substantially reduced in injured corneas of CX3CR1-deficient mice compared with WT mice, consistent with previous reports that Ab-mediated inhibition of CX3CL1 reduced macrophage migration into the affected organs in experimental allergic myositis and collagen-induced arthritis (46, 47). Thus, the CXC3CL1-CX3CR1 interactions may have an important role in macrophage trafficking. However, CX3CR1 deficiency failed to abrogate completely macrophage accumulation into injured cornea, suggesting a contribution of other chemokines and/or other chemotactic factors to macrophage accumulation. Indeed, we observed that alkali injury enhanced intraocular mRNA expression of CCL2 and CCL3 (data not shown), the chemokines that exhibit potent chemotactic activities for macrophages. Thus, it is likely that alkali-induced macrophage accumulation is regulated by the combined action of CX3CL1 with other macrophage-tropic chemokines such as CCL2 and CCL3.

Macrophages are presumed to be proangiogenic in various types of pathological conditions (14, 17, 36, 37). Selective macrophage depletion with clodronate liposomes inhibited laser photocoagulation-induced VEGF production and subsequent choroidal neovascularization (13). Furthermore, Ambati et al. (19, 20) reported that CNV and VEGF production was suppressed in mice lacking CCR2 or CCR5, the specific chemokine receptors for CCL2 and CCL3, respectively. Thus, CCR2- and/or CCR5-expressing macrophages can promote neovascularization, probably by producing VEGF. On the contrary, under our present experimental conditions, genetic depletion of CX3CR1 reduced alkali injury-induced macrophage accumulation but promoted CNV with little effects on intraocular VEGF expression. Similarly, Apte et al. (21) demonstrated that inhibition of macrophage accumulation promoted laser photocoagulation-induced choroidal neovascularization, whereas direct injection of macrophages suppressed it (21). Considering the proposed hypothesis on the dichotomy of macrophages based on the expression levels of CCR2 and CX3CR1 (26, 27, 28, 29, 30), CX3CR1-expressing macrophages may exhibit biological functions distinct from CCR2-expressing ones, in the course of angiogenesis.

Normal cornea lacks any vasculature under physiological conditions. This physiological corneal avascularity is presumed to be maintained by the balance between angiogenic and antiangiogenic factor expression (48, 49, 50, 51, 52). Soluble VEGF receptor 1 is presumed to be a major antiangiogenic factor responsible for the maintenance of this physiological corneal avascularity by trapping constitutively expressed VEGF (53). However, it still remains to be established whether soluble VEGF receptor 1 can counteract CNV induced by corneal injury. Although CX3CR1-deficient mice exhibited intracorneal VEGF mRNA expression to a similar extent as WT mice, they still developed a more severe form of CNV than WT mice. Thus, the VEGF/soluble VEGF receptor 1 interactions may not be responsible for enhanced CNV observed in CX3CR1-deficient mice.

Alternatively, antiangiogenic molecules, TSP-1 and TSP-2, which are also constitutively present in cornea, can preclude vasculature development (48, 54, 55). TSP-1 levels in Bruch’s membrane and choroidal vessels was decreased in age-related macular degeneration (AMD) and a reduction in TSP-1 may permit the formation of choroidal neovascularization (56). Moreover, TSP-1 deficiency and, to a lesser extent, TSP-2 deficiency resulted in enhanced CNV induced by suture placement (48). TSP-1 and TSP-2 are processed to release antiangiogenic polypeptides by the action of ADAMTS-1 (57). ADAMTS-1 at a high concentration can also inhibit endothelial migration induced by combined treatment with VEGF and basic fibroblast growth factor (38). Mirroring its antiangiogenic activities, ADAMTS-1-deficient mice exhibited delayed skin wound closure, along with exaggerated neovascularization (57). Because the enhanced expression of TSP-1, TSP-2, and ADAMTS-1 was attenuated in CX3CR1-deficient mice following alkali injury, the reduced expression of these factors may be responsible for enhanced alkali-induced CNV in CX3CR1-deficient mice.

Monocyte/macrophages are a major source of TSP-1 (58). ADAMTS-1 was also detected in infiltrating macrophages in the early phase of skin wound sites (38). TSP-1 and ADAMTS-1 proteins were consistently detected in F4/80-positive and CX3CR1-positive cell populations in this model of alkali-induced cornea injury. Moreover, CX3CL1 augmented TSP-1 and ADAMTS-1 expression by murine peritoneal macrophages. Thus, potent antiangiogenic molecules, TSP-1 and ADAMTS-1, may be expressed by CX3CR1-positve macrophages. This may account for protective roles of the CX3CL1-CX3CR1 interactions in alkali injury-induced CNV.

Chemical burns of the eyes can destroy surface epithelia and cause ischemic necrosis of eye tissues and, if not treated properly, CNV ensues and impairs vision. Alkali, once applied onto eyes, is difficult to be completely rinsed away due to its rapid penetration into the anterior chamber and, therefore, an effective maneuver(s) is required to prevent alkali injury-induced CNV. Glucocorticoids are widely used for alkali injury of eyes with limited therapeutic benefits (59, 60). In this study, we demonstrated that topical application of CX3CL1 markedly augmented the intraocular expression of antiangiogenic molecules, TSP-1 and ADAMTS-1, without affecting VEGF expression. Moreover, topical application of CX3CL1, even when started after the injury, was very effective in preventing alkali injury-induced CNV. These observations raise the possibility of CX3CL1 as a preventive drug for alkali injury-induced CNV.

Two single nucleotide polymorphisms within the CX3CR1 gene were reported to be associated with AMD and reduce CX3CR1 protein expression (61, 62). Because choroidal neovascularization is a characteristic pathological feature of AMD, depressed CX3CR1 expression may be responsible for neovascularization in AMD, as observed in alkali-induced CNV. If so, topical application of CX3CL1 may be effective for the prevention and treatment of AMD, which is the leading cause of visual impairment and blindness among people age 65 years and older.

We thank Drs. Philip M. Murphy and Ji-Liang Gao (National Institute of Allergy and Infectious Diseases, National Institutes of Health) for providing us with CX3CR1-deficient mice and Drs. Masanobu Oshima and Hiroko Oshima (Division of Genetics, Cancer Research Institute, Kanazawa University) and Dr. Toshikazu Kondo (Department of Forensic Medicine, Wakayama Medical University, Wakayama City, Japan) for their technical assistance. We express our thanks to Dr. Joost J. Oppenheim (National Cancer Institute, Frederick, MD) for his critical reading of this manuscript.

The authors have no financial conflict of interest.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1

This work was supported by the International Cooperative Program of Kanazawa University (to N.M.) and the National Natural Science Foundation in China (Grants 30572120 and 30771978), the Jiangsu Natural Science Foundation (Grant BK2006528), the China Postdoctoral Science Foundation (Grant 2005038587), grants from the Suzhou University (to P.L.), and the National Natural Science Key Program Foundation in China (Grant 30330540 to X.Z.).

3

Abbreviations used in this paper: CNV, corneal neovascularization; ADAMTS, a disintegrin and metalloprotease with thrombospondin; pAb, polyclonal Ab; VEGF, vascular endothelial growth factor; TSP, thrombospondin; WT, wild type; CT, threshold cycle; AMD, age-related macular degeneration.

1
Chang, J. H., E. E. Gabison, T. Kato, D. T. Azar.
2001
. Corneal neovascularization.
Curr. Opin. Ophthalmol.
12
:
242
-249.
2
Epstein, R. J., R. D. Stulting, R. L. Hendricks, D. M. Harris.
1987
. Corneal neovascularization: pathogenesis and inhibition.
Cornea
6
:
250
-257.
3
Klintworth, G. K., P. C. Burger.
1983
. Neovascularization of the cornea: current concepts of its pathogenesis.
Int. Ophthalmol. Clin.
23
:
27
-39.
4
Cursiefen, C., J. Cao, L. Chen, Y. Liu, K. Maruyama, D. Jackson, F. E. Kruse, S. J. Wiegand, M. R. Dana, J. W. Streilein.
2004
. Inhibition of hemangiogenesis and lymphangiogenesis after normal-risk corneal transplantation by neutralizing VEGF promotes graft survival.
Invest. Ophthalmol. Visual Sci.
45
:
2666
-2673.
5
Streilein, J. W., J. Yamada, M. R. Dana, B. R. Ksander.
1999
. Anterior chamber-associated immune deviation, ocular immune privilege, and orthotopic corneal allografts.
Transplant. Proc.
31
:
1472
-1475.
6
Dana, M. R., J. W. Streilein.
1996
. Loss and restoration of immune privilege in eyes with corneal neovascularization.
Invest. Ophthalmol. Visual Sci.
37
:
2485
-2494.
7
Sano, Y., B. R. Ksander, J. W. Streilein.
1995
. Fate of orthotopic corneal allografts in eyes that cannot support anterior chamber-associated immune deviation induction.
Invest. Ophthalmol. Visual Sci.
36
:
2176
-2185.
8
Streilein, J. W., D. Bradley, Y. Sano, Y. Sonoda.
1996
. Immunosuppressive properties of tissues obtained from eyes with experimentally manipulated corneas.
Invest. Ophthalmol. Visual Sci.
37
:
413
-424.
9
Cursiefen, C., K. Maruyama, D. G. Jackson, J. W. Streilein, F. E. Kruse.
2006
. Time course of angiogenesis and lymphangiogenesis after brief corneal inflammation.
Cornea
25
:
443
-447.
10
Lu, P., L. Li, N. Mukaida, X. Zhang.
2007
. Alkali-induced corneal neovascularization is independent of CXCR2-mediated neutrophil infiltration.
Cornea
26
:
199
-206.
11
Zhou, J., L. Pham, N. Zhang, S. He, M. A. Gamulescu, C. Spee, S. J. Ryan, D. R. Hinton.
2005
. Neutrophils promote experimental choroidal neovascularization.
Mol. Vis.
11
:
414
-424.
12
Tsutsumi-Miyahara, C., K. H. Sonoda, K. Egashira, M. Ishibashi, H. Qiao, T. Oshima, T. Murata, M. Miyazaki, I. F. Charo, S. Hamano, T. Ishibashi.
2004
. The relative contributions of each subset of ocular infiltrated cells in experimental choroidal neovascularisation.
Br. J. Ophthalmol.
88
:
1217
-1222.
13
Taub, D. D..
1996
. Chemokine-leukocyte interactions: the voodoo that they do so well.
Cytokine Growth Factor Rev.
7
:
355
-376.
14
Sakurai, E., A. Anand, B. K. Ambati, N. van Rooijen, J. Ambati.
2003
. Macrophage depletion inhibits experimental choroidal neovascularization.
Invest. Ophthalmol. Visual Sci.
44
:
3578
-3585.
15
Sakurai, E., H. Taguchi, A. Anand, B. K. Ambati, E. S. Gragoudas, J. W. Miller, A. P. Adamis, J. Ambati.
2003
. Targeted disruption of the CD18 or ICAM-1 gene inhibits choroidal neovascularization.
Invest. Ophthalmol. Visual Sci.
44
:
2743
-2749.
16
Grossniklaus, H. E., J. X. Ling, T. M. Wallace, S. Dithmar, D. H. Lawson, C. Cohen, V. M. Elner, S. G. Elner, P. Sternberg.
2002
. Macrophage and retinal pigment epithelium expression of angiogenic cytokines in choroidal neovascularization.
Mol. Vis.
21
:
119
-126.
17
Tsutsumi, C., K. H. Sonoda, K. Egashira, H. Qiao, T. Hisatomi, S. Nakao, M. Ishibashi, I. F. Charo, T. Sakamoto, T. Murata, T. Ishibashi.
2003
. The critical role of ocular-infiltrating macrophages in the development of choroidal neovascularization.
J. Leukocyte Biol.
74
:
25
-32.
18
Ambati, B. K., A. M. Joussen, W. A. Kuziel, A. P. Adamis, J. Ambati.
2003
. Inhibition of corneal neovascularization by genetic ablation of CCR2.
Cornea
22
:
465
-467.
19
Ambati, B. K., A. Anand, A. W. Joussen, W. A. Kuziel, A. P. Adamis, J. Ambati.
2003
. Sustained inhibition of corneal neovascularization by genetic ablation of CCR5.
Invest. Ophthalmol. Visual Sci.
44
:
590
-593.
20
Ambati, J., A. Anand, S. Fernandez, E. Sakurai, B. C. Lynn, W. A. Kuziel, B. J. Rollins, B. K. Ambati.
2003
. An animal model of age-related macular degeneration in senescent Ccl-2- or Ccr-2-deficient mice.
Nat. Med.
9
:
1390
-1397.
21
Apte, R. S., J. Richter, J. Herndon, T. A. Ferguson.
2006
. Macrophages inhibit neovascularization in a murine model of age-related macular degeneration.
PLoS Med.
3
:
1371
-1381.
22
Furuichi, K., J. L. Gao, P. M. Murphy.
2006
. Chemokine receptor CX3CR1 regulates renal interstitial fibrosis after ischemia-reperfusion injury.
Am. J. Pathol.
169
:
372
-387.
23
Barlic, J., Y. Zhang, J. F. Foley, P. M. Murphy.
2006
. Oxidized lipid-driven chemokine receptor switch, CCR2 to CX3CR1, mediates adhesion of human macrophages to coronary artery smooth muscle cells through a peroxisome proliferator-activated receptor γ-dependent pathway.
Circulation
114
:
807
-819.
24
Imaizumi, T., H. Yoshida, K. Satoh.
2004
. Regulation of CX3CL1/fractalkine expression in endothelial cells.
J. Atheroscler. Thromb.
11
:
15
-21.
25
Combadiere, C., S. Potteaux, J. L. Gao, B. Esposito, S. Casanova, E. J. Lee, P. Debre, A. Tedgui, P. M. Murphy, Z. Mallat.
2003
. Decreased atherosclerotic lesion formation in CX3CR1/apolipoprotein E double knockout mice.
Circulation
107
:
1009
-1016.
26
Mantovani, A., A. Sica, S. Sozzani, P. Allavena, A. Vecchi, M. Locati.
2004
. The chemokine system in diverse forms of macrophage activation and polarization.
Trends Immunol.
25
:
677
-686.
27
Landsman, L., C. Varol, S. Jung.
2007
. Distinct differentiation potential of blood monocyte subsets in the lung.
J. Immunol.
178
:
2000
-2007.
28
Yrlid, U., C. D. Jenkins, G. G. MacPherson.
2006
. Relationships between distinct blood monocyte subsets and migrating intestinal lymph dendritic cells in vivo under steady-state conditions.
J. Immunol.
176
:
4155
-4162.
29
Tacke, F., D. Alvarez, T. J. Kaplan, C. Jakubzick, R. Spanbroek, J. Llodra, A. Garin, J. Liu, M. Mack, N. van Rooijen, et al
2007
. Monocyte subsets differentially employ CCR2, CCR5, and CX3CR1 to accumulate within atherosclerotic plaques.
J. Clin. Invest.
117
:
185
-194.
30
Geissmann, F., S. Jung, D. R. Littman.
2003
. Blood monocytes consist of two principal subsets with distinct migratory properties.
Immunity
19
:
71
-82.
31
Yang, X., P. Lu, C. Fujii, Y. Nakamoto, J. L. Gao, S. Kaneko, P. M. Murphy, N. Mukaida. Essential contribution of a chemokine, CCL3, and its receptor, CCR1, to hepatocellular carcinoma progression.
Int. J. Cancer
118
:
1869
-1876.
32
Yang, X., P. Lu, Y. Ishida, W. A. Kuziel, C. Fujii, N. Mukaida.
2006
. Attenuated liver tumor formation in the absence of CCR2 with a concomitant reduction in the accumulation of hepatic stellate cells, macrophages and neovascularization.
Int. J. Cancer
118
:
335
-345.
33
Oshima, T., K. H. Sonoda, C. Tsutsumi-Miyahara, H. Qiao, T. Hisatomi, S. Nakao, S. Hamano, K. Egashira, I. F. Charo, T. Ishibashi.
2006
. Analysis of corneal inflammation induced by cauterisation in CCR2 and MCP-1 knockout mice.
Br. J. Ophthalmol.
90
:
218
-222.
34
Nakano, Y., T. Kasahara, N. Mukaida, Y. C. Ko, M. Nakano, K. Matsushima.
1994
. Protection against lethal bacterial infection in mice by monocyte-chemotactic and -activating factor.
Infect. Immun.
62
:
377
-383.
35
Lu, P., Y. Nakamoto, Y. Nemoto-Sasaki, C. Fujii, H. Wang, M. Hashii, Y. Ohmoto, S. Kaneko, K. Kobayashi, N. Mukaida.
2003
. Potential interaction between CCR1 and its ligand, CCL3, induced by endogenously produced interleukin-1 in human hepatomas.
Am. J. Pathol.
162
:
1249
-1258.
36
Espinosa-Heidmann, D. G., I. J. Suner, E. P. Hernandez, D. Monroy, K. G. Csaky, S. W. Cousins.
2003
. Macrophage depletion diminishes lesion size and severity in experimental choroidal neovascularization.
Invest. Ophthalmol. Visual Sci.
44
:
3586
-3592.
37
Oh, H., H. Takagi, C. Takagi, K. Suzuma, A. Otani, K. Ishida, M. Matsumura, Y. Ogura, Y. Honda.
1999
. The potential angiogenic role of macrophages in the formation of choroidal neovascular membranes.
Invest. Ophthalmol. Visual Sci.
40
:
1891
-1898.
38
Krampert, M., S. Kuenzle, S. N. Thai, N. Lee, M. L. Iruela-Arispe, S. Werner.
2005
. ADAMTS1 proteinase is up-regulated in wounded skin and regulates migration of fibroblasts and endothelial cells.
J. Biol. Chem.
280
:
23844
-23852.
39
Benelli, R., A. Albini, D. Noonan.
2003
. Neutrophils and angiogenesis: potential initiators of the angiogenic cascade.
Chem. Immunol. Allergy
83
:
167
-181.
40
Gaudry, M., O. Bregerie, V. Andrieu, J. El Benna, M. A. Pocidalo, J. Hakim.
1997
. Intracellular pool of vascular endothelial growth factor in human neutrophils.
Blood
90
:
4153
-4161.
41
McCourt, M., J. H. Wang, S. Sookhai, R. P. Redmond.
2001
. Activated human neutrophils release hepatocyte growth factor/scatter factor.
Eur. J. Surg. Oncol.
27
:
396
-403.
42
Ohki, Y., B. Heissig, Y. Sato, H. Akiyama, Z. Zhu, D. J. Hicklin, K. Shimada, H. Ogawa, H. Daida, K. Hattori, A. Ohsaka.
2005
. Granulocyte colony-stimulating factor promotes neovascularization by releasing vascular endothelial growth factor from neutrophils.
FASEB J.
19
:
2005
-2007.
43
Morini, M., C. Tecchio, S. Minghelli, E. Di Carlo, E. Tanghetti, A. Albini, C. Lowell, G. Berton, D. M. Noonan, M. A. Cassatella.
2004
. CXCL1/macrophage inflammatory protein-2-induced angiogenesis in vivo is mediated by neutrophil-derived vascular endothelial growth factor-A.
J. Immunol.
172
:
5034
-5040.
44
Scapini, P., F. Calzetti, M. A. Cassatella.
1999
. On the detection of neutrophil-derived vascular endothelial growth factor (VEGF).
J. Immunol. Methods
232
:
121
-129.
45
Liu, G. Y., V. Kulasingam, R. T. Alexander, N. Touret, A. M. Fong, D. D. Patel, L. A. Robinson.
2005
. Recycling of the membrane-anchored chemokine, CX3CL1.
J. Biol. Chem.
280
:
19858
-19866.
46
Suzuki, F., T. Nanki, T. Imai, H. Kikuchi, S. Hirohata, H. Kohsaka, N. Miyasaka.
2005
. Inhibition of CX3CL1 (fractalkine) improves experimental autoimmune myositis in SJL/J mice.
J. Immunol.
175
:
6987
-6996.
47
Nanki, T., Y. Urasaki, T. Imai, M. Nishimura, K. Muramoto, T. Kubota, N. Miyasaka.
2004
. Inhibition of fractalkine ameliorates murine collagen-induced arthritis.
J. Immunol.
173
:
7010
-7016.
48
Cursiefen, C., S. Masli, T. F. Ng, M. R. Dana, P. Bornstein, J. Lawler, J. W. Streilein.
2004
. Roles of thrombospondin-1 and -2 in regulating corneal and iris angiogenesis.
Invest. Ophthalmol. Visual Sci.
45
:
1117
-1124.
49
Gao, G., Y. Li, D. Zhang, S. Gee, C. Crosson, J. Ma.
2001
. Unbalanced expression of VEGF and PEDF in ischemia-induced retinal neovascularization.
FEBS Lett.
489
:
270
-276.
50
Gao, G., J. Ma.
2002
. Tipping the balance for angiogenic disorders.
Drug Discov. Today
7
:
171
-172.
51
Zhang, S. X., J. X. Ma.
2007
. Ocular neovascularization: implication of endogenous angiogenic inhibitors and potential therapy.
Prog. Retin. Eye Res.
26
:
1
-37.
52
Dorrell, M. I., E. Aguilar, L. Scheppke, F. H. Barnett, M. Friedlander.
2007
. Combination angiostatic therapy completely inhibits ocular and tumor angiogenesis.
Proc. Natl. Acad. Sci. USA
104
:
967
-972.
53
Ambati, B. K., M. Nozaki, N. Singh, A. Takeda, P. D. Jani, T. Suthar, R. J. C. Albuquerque, E. Richter, E. Sakurai, M. T. Newcomb, et al
2006
. Corneal avascularity is due to soluble VEGF receptor-1.
Nature
443
:
993
-997.
54
Choudhary, A., P. Hiscott, C. A. Hart, S. B. Kaye, M. Batterbury, I. Grierson.
2005
. Suppression of thrombospondin 1 and 2 production by herpes simplex virus 1 infection in cultured keratocytes.
Mol. Vis.
11
:
163
-168.
55
Uno, K., H. Hayashi, M. Kuroki, H. Uchida, Y. Yamauchi, M. Kuroki, K. Oshima.
2004
. Thrombospondin-1 accelerates wound healing of corneal epithelia.
Biochem. Biophys. Res. Commun.
315
:
928
-934.
56
Uno, K., I. A. Bhutto, D. S. McLeod, C. Merges, G. A. Lutty.
2006
. Impaired expression of thrombospondin-1 in eyes with age related macular degeneration.
Br. J. Ophthalmol.
90
:
48
-54.
57
Lee, N. V., M. Sato, D. S. Annis, J. A. Loo, L. Wu, D. F. Mosher, M. L. Iruela-Arispe.
2006
. ADAMTS1 mediates the release of antiangiogenic polypeptides from TSP1 and 2.
EMBO J.
25
:
5270
-5283.
58
Yesner, L. M., H. Y. Huh, S. F. Pearce, R. L. Silverstein.
1996
. Regulation of monocyte CD36 and thrombospondin-1 expression by soluble mediators.
Arterioscler. Thromb. Vasc. Biol.
16
:
1019
-1025.
59
Sekundo, W., A. J. Augustin, I. Strempel.
2002
. Topical allopurinol or corticosteroids and acetylcysteine in the early treatment of experimental corneal alkali burns: a pilot study.
Eur. J. Ophthalmol.
12
:
366
-372.
60
Den, S., C. Sotozono, S. Kinoshita, T. Ikeda.
2004
. Efficacy of early systemic β-methasone or cyclosporin A after corneal alkali injury via inflammatory cytokine reduction.
Acta Ophthalmol. Scand.
82
:
195
-199.
61
Chan, C. C., J. Tuo, C. M. Bojanowski, K. G. Csaky, W. R. Green.
2005
. Detection of CX3CR1 single nucleotide polymorphism and expression on archived eyes with age-related macular degeneration.
Histol. Histopathol.
20
:
857
-863.
62
Tuo, J., B. C. Smith, C. M. Bojanowski, A. D. Meleth, I. Gery, K. G. Csaky, E. Y. Chew, C. C. Chan.
2004
. The involvement of sequence variation and expression of CX3CR1 in the pathogenesis of age-related macular degeneration.
FASEB J.
18
:
1297
-1299.