Introduction: This study aimed to investigate the characteristics of retinal vascular degeneration and the expression of vessel-related claudin (CLD) proteins in retinal degeneration mouse (Pde6βrd1/rd1 rd1 mouse). Methods: Retinas from wild-type (WT) mice and rd1 mice at postnatal day 3 (P3), P5, P8, P11, P13, P15, P18, and P21 were collected. Immunofluorescence staining was used to assess the retinal vascular plexus, cell proliferation, CLD expression, and retinal ganglion cells (RGCs). The distribution of retinal superficial and deep vessels was determined by isolectin B4 fluorescence staining of retinal flat mounts and frozen sections. Hematoxylin and eosin staining and terminal deoxynucleotidyl transferase-mediated dNTP nick-end labeling were used to investigate retinal histological degeneration and apoptosis in rd1 mice, respectively. Quantitative real-time PCR and Western blot were used to measure the expression of vessel-related CLD-1, -2, -3, and -5, vascular endothelial growth factor A (VEGFA), and vascular endothelial growth factor receptor 2 (VEGFR2) in the retinas. Results: Compared to the WT mice, the rd1 mice displayed delayed but completed progressive development in the retinal superficial vascular plexuses (SVPs) and deep vascular plexuses (DVPs). In the rd1 mice, the thickness of retinal layers gradually decreased and the retinas underwent progressive atrophy and degeneration. The deterioration got worse at the late developmental stage. The declined vessel density of SVP and DVP correlated with the decreased thickness of the full and inner parts of the retina and the reduced number of RGCs. DVP degeneration and the thinning of the outer nuclear layer exhibited an obvious reduction at P15. The expression levels of CLD-1, CLD-2, CLD-3, CLD-5, VEGFA, and VEGFR2 decreased and were consistently lower in the rd1 mice than in WT mice since P15. Conclusion: Rd1 mice exhibited progressive vascular degeneration of retinal SVP and DVP, the thinning and atrophy of retinal ONL and RGC, and the downregulation of vessel-related CLD proteins during the late developmental period. Thus, the rd1 mouse is a useful model of not only retinal neuro-degeneration but also retinal vascular degeneration.

Retinal degeneration models are widely used to investigate the pathogenesis of retinitis pigmentosa (RP) in humans. This hereditary retinopathy is characterized by the progressive loss of retinal photoreceptor cells and visual acuity and the eventual blindness [1, 2]. As retinal photoreceptors undergo apoptosis, the metabolic demand of the photoreceptor cells decreases and the ocular microcirculation is altered in RP [3‒6]. The decreased blood flow of retinal and choroidal vessels in patients with RP has been found in some studies [4‒8]. However, the mechanisms of retinal vascular alteration and its association with photoreceptor degeneration need to be further clarified.

Claudins (CLDs) are 20–27 kDa proteins with over 24 isoforms in humans and mice [9, 10]. CLDs are crucial proteins primarily localized at the intercellular junctions of epithelial cells, endothelial cells (ECs), and tumor cells. CLDs can regulate the intercellular transport of water and ions and play a vital role in maintaining cell polarity, cell proliferation, barrier function, and cell signaling [11, 12]. Our previous studies have demonstrated that CLD-1, -2, and -5 specifically express on retinal vessels; and the overexpression of CLD-5 can decrease cell mobility and the sprouting capability of the vessel in vitro and induce new interaction patterns between CLD-5 and CLD-1 or -2 in retinal pigment epithelium cells and human retina ECs [13, 14]. CLD-1, -2, and -5 are implicated in the development and maintenance of the blood-retinal barrier (BRB), as well as the preservation of ECs’ structural integrity [13, 15‒17]. The expression of CLD-5 upregulated by JAM-A, a component of tight junctions, can reduce endothelial permeability [18]. Notably, CLD-3 is the sole known isoform of CLD that interacts with CLD-1, -2, and -5 through extracellular loops and then forms heterodimers [19]. It has been proved that elevated intraocular pressure leads to a significant decrease in the expression of CLD-3 and CLD-5, along with the up-regulation of interleukin 1-β and eNOS as well as the injury of BRB [20]. Our previous studies have also found that CLD-3 mainly locates at the retinal ganglion cells (RGCs) and the downregulation of claudin-3 can impede retinal vessel development in mice [13, 21], and CLD-3 is also required for normal development of the neural retina and retinal vessels in zebrafish [22]. Therefore, CLD-3 can also be considered a vessel-related CLD. These findings collectively unveil the importance of CLDs that can affect the development and stabilization of retinal vessels and retinal nerves.

The trait of rd1 (Pde6βrd1/rd1) mice is caused by a recessive mutation of Pde6β, which encodes the rod β-subunit of cyclic guanosine monophosphate phosphodiesterase [23, 24]. The photoreceptor development is initially normal in rd1 mouse, but all rod photoreceptors start to degenerate at the postnatal 2 weeks; then the cone photoreceptors gradually degenerate [23]. As the rd1 mouse shares the pathophysiological features with RP disease, the rd1 mouse is a valuable tool for studying RP [25]. The study aimed to investigate the characteristics of retinal vascular degeneration and the expression of vessel-related CLDs and analyze the correlations between the degeneration of retinal vasculature and retinal neurons in rd1 mice.

Animals

All animal experiments adhered to the ARVO Statement for the use of animals in ophthalmic and vision research and were approved by the Institutional Review Board of Zhongshan Ophthalmic Center (Guangzhou, China). The rd1 mice (FVB/rd1) and C57BL/6 wild-type (WT) mice were purchased, respectively, from Vital River Laboratory Animal Technology Co. Ltd. (Beijing, China) and Southern Medical University (Guangzhou, China). All animals were kept in specific pathogen-free animal laboratory of Zhongshan Ophthalmic Center (Guangzhou, China) with standard feeding conditions. The retinas of two groups were collected at postnatal day 3 (P3), P5, P8, P11, P13, P15, P18, and P21 for the following experiments.

Fluorescence Staining of Retinal Flatmount and Retinal Frozen Section

To evaluate retinal vascularization and vascular distribution, retinal flatmounts and retinal frozen section were determined by isolectin B4 (IB4) fluorescence staining at P3, P5, P8, P11, P13, P15, P18, and P21 as previously described [21, 26, 27]. After cardiac perfusion with 30 mL phosphate-buffered saline and 20–30 mL 4% (v/v) paraformaldehyde, the eyes were enucleated and fixed in 4% (v/v) paraformaldehyde for 45 min at 4°C, then dissected to make retinal flatmounts and retinal frozen section. The retinal flatmounts were permeabilized with 0.5% Triton X-100 in phosphate-buffered saline for 10 min, then blocked with 20% (v/v) fetal bovine serum (Gibco, USA) for 1 h at room temperature, and then stained with vascular marker Alexa 568-conjugated isolectin B4 (1:200, Invitrogen, Carlsbad, CA, USA) and anti-ki67 primary antibody (1:100, Cell Signaling Technology [CST], Danvers, USA) to detect proliferation at 4°C overnight [26]. The retinas were then labeled with Alexa Fluor® 488-conjugated secondary antibody (1:1,000, CST) for 1 h at room temperature. Vascular distribution and tip cells were identified under a confocal microscope (LSM 980, Carl Zeiss AG, Oberkochen, Germany). Adobe Photoshop (Adobe, Inc., San Jose, CA, USA) and ImageJ (National Institute of Health [NIH], Bethesda, MD, USA) were used to analyze the retinal vasculature.

Immunofluorescence Staining of Retinal Frozen Section

Mouse eyes enucleated at P3, P5, P8, P11, P13, P15, P18, and P21 were fixed and embedded in optimal cutting temperature compound (Sakura Finetek USA, Inc., Torrance, CA, USA) at −80°C and sectioned as previously described [26, 27]. The retinal cryosections were stained with Alexa® 568-conjugated IB4 (1:200), anti-CLD-2 antibody (1:250, Abcam, Cambridge, MA, USA), anti-CLD-3 antibody (1:100, Invitrogen), and anti-RBPMS antibody (1:200, GeneTex, Inc., Irvine, CA, USA) to detect the RGCs at 4°C overnight. The retinas were then probed with anti-rabbit Alexa Fluor 488 secondary antibody (1:1,000, CST, Danvers, MA, USA) and then incubated with 4′,6-diamidino-2-phenylindole (1:2,000, Invitrogen) for 15 min to visualize the cell nuclei as previously described [27]. Sections were viewed under a confocal microscope. ImageJ software was used to examine the distributions of the retinal vessels.

Western Blot

Retinal proteins at P3, P5, P11, P15, P18, and P21 were extracted by radioimmunoprecipitation assay buffer (Beyotime, Shanghai, China) with protease (Absin, Shanghai, China) and phosphatase inhibitors (Absin, Shanghai, China). The protein concentration was measured by Pierce BCA Protein Assay Kit (Thermo Fisher Scientific, USA) according to the manufacturer’s instructions. Western blotting was performed on SurePAGE and Bis-Tris gels (GenScript Biotech Corp., Biotech Corp., Piscataway, NJ, USA). The primary antibodies used were anti-β-actin (1:5,000), anti-VEGFA (1:1,000), anti-VEGFR2 (1:1,000) (all from CST), anti-CLD-1 (1:1,000) (HUABIO, Woburn, MA, USA), anti-CLD-2 (1:1,000, Abcam), anti-CLD-3 (1:1,000) (Invitrogen, USA), and anti-CLD-5 (1:1,000, Abcam). Anti-rabbit IgG HRP-linked antibodies (1:20,000, CST) were used to incubate membranes. Signals were detected with ChemiDoc MP Imaging System (BioRad, USA). Quantification of the protein expression was measured by ImageJ. All experimental steps were conducted as previously described [13, 26, 27].

Quantitative Real-Time PCR

Retinal RNA at P3, P5, P8, P11, P13, P15, P18, and P21 was extracted with TRIzol reagent (Invitrogen) and synthesized the cDNA with a cDNA Synthesis Kit (TaKaRa Bio Inc., Kusatsu, Shiga, Japan). Synthesis of cDNA and RT-qPCR was performed as previously described [27, 28]. Information of the PCR primers was summarized in Table 1. GAPDH was used as the reference gene to normalize the total mRNA levels.

Table 1.

Primers used in quantitative real-time PCR

GenesSequencing 5′ to 3′
Mouse GAPDH F AGG​TCG​GTG​TGA​ACG​GAT​TTG 
Mouse GAPDH R TGT​AGA​CCA​TGT​AGT​TGA​GGT​C 
Mouse claudin 1 F TCT​ACG​AGG​GAC​TGT​GGA​TG 
Mouse claudin 1 R TCA​GAT​TCA​GCT​AGG​AGT​CG 
Mouse claudin 2 F GGC​TGT​TAG​GCT​CAT​CCA​T 
Mouse claudin 2 R TGG​CAC​CAA​CAT​AGG​AAC​TC 
Mouse claudin 3 F AAG​CCG​AAT​GGA​CAA​AGA​A 
Mouse claudin 3 R CTG​GCA​AGT​AGC​TGC​AGT​G 
Mouse claudin 5 F GTG​GAA​CGC​TCA​GAT​TTC​AT 
Mouse claudin 5 R TGG​ACA​TTA​AGG​CAG​CAT​CT 
GenesSequencing 5′ to 3′
Mouse GAPDH F AGG​TCG​GTG​TGA​ACG​GAT​TTG 
Mouse GAPDH R TGT​AGA​CCA​TGT​AGT​TGA​GGT​C 
Mouse claudin 1 F TCT​ACG​AGG​GAC​TGT​GGA​TG 
Mouse claudin 1 R TCA​GAT​TCA​GCT​AGG​AGT​CG 
Mouse claudin 2 F GGC​TGT​TAG​GCT​CAT​CCA​T 
Mouse claudin 2 R TGG​CAC​CAA​CAT​AGG​AAC​TC 
Mouse claudin 3 F AAG​CCG​AAT​GGA​CAA​AGA​A 
Mouse claudin 3 R CTG​GCA​AGT​AGC​TGC​AGT​G 
Mouse claudin 5 F GTG​GAA​CGC​TCA​GAT​TTC​AT 
Mouse claudin 5 R TGG​ACA​TTA​AGG​CAG​CAT​CT 

Hematoxylin and Eosin Staining of Retinal Paraffin Sections

Eyeballs from rd1 and WT mice at P5, P8, P11, P15, P18, and P21 were fixed in FAS eyeball fixative solution (Servicebio, Technology Co. Ltd., Wuhan, China). Eye cups were embedded in paraffin, dehydrated with ethanol and xylene, and sectioned. The retinal paraffin sections were stained with hematoxylin and eosin and sealed with neutral glue as previously described [27]. HE-stained sections were used to analyze the retinal thickness of the regions, which is 350 μm adjacent to the optic nerve. The thickness of the inner part of the retina (IPR) and outer nuclear layer (ONL) and the number of RGCs were measured at the same region [29‒32]. IPR included the outer plexiform layer, inner nuclear layer, inner plexiform layer, ganglion cell layer, and nerve fiber layer (NFL) [30]. Images were scanned and measured with TissueFAXS (TissueGnostics Asia Pacific, Beijing, China).

TdT-Mediated dUTP Nick-End Labeling Staining

Cell apoptosis was quantified in retinal frozen sections of P8, P11, P15, P18, P21 rd1 mice and WT mice using TdT-mediated dUTP nick-end labeling (TUNEL) In Situ Cell Death Detection Kit (Roche Diagnostics, Risch-Rotkreuz, Switzerland) as previously described [21].

Statistical Analysis

Two independent masked investigators performed all counting and measurements in duplicate. All data are expressed as means ± standard error of the mean. Each experiment was repeated at least three times. The data were analyzed and the graphs were plotted with GraphPad Prism v. 8.0 (GraphPad Software, La Jolla, CA, USA), ImageJ (NIH), and SPSS v. 23.0 (IBM Corp., Armonk, NY, USA). One-way analysis of variance and Spearman’s correlation coefficients were calculated. A p value <0.05 was considered statistically significant.

The Development of Retinal Superficial Vascular Plexus and Deep Vascular Plexus in Rd1 Mice Lags behind Those in WT Mice

There was no significant difference in the Ki67-positive ECs between rd1 mice and WT mice at P3, which is the early developmental stage of superficial vascular plexus (SVP). SVP of both groups was extremely active and growing toward the peripheral retina, and overall growth progress of SVP was slower in rd1 mice compared to WT mice. The WT mice had more Ki67-positive ECs than the rd1 mice at P5. At P8, the SVP termini formed a closed loop in WT mice; however, the rd1 mice had more Ki67-positive ECs than WT mice (Fig. 1a, c).

Fig. 1.

Superficial vascular plexus (SVP) at postnatal day 3 (P3) to P8. a The retinal flatmounts stained with isolectin B4 (IB4) (red) and anti-Ki67 antibody (green) were detected. Scale bar, 100 μm. b Representative images of retinal flatmounts and tip cells were marked out. Scale bar, 100 μm. c Quantitative analysis of the number of Ki67-positive ECs between WT mice and rd1 mice, showing statistical differences at P5 and P8 (n = 6). d Statistical analysis of superficial vascular density showed a statistical difference during P3 to P8 between WT mice and rd1 mice (n = 6). e The number of tip cells was statistically different at P5 and P8 between WT mice and rd1 mice (n = 6). The data are presented as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. WT, wild type; IB4, isolectin B4; ECs, endothelial cells.

Fig. 1.

Superficial vascular plexus (SVP) at postnatal day 3 (P3) to P8. a The retinal flatmounts stained with isolectin B4 (IB4) (red) and anti-Ki67 antibody (green) were detected. Scale bar, 100 μm. b Representative images of retinal flatmounts and tip cells were marked out. Scale bar, 100 μm. c Quantitative analysis of the number of Ki67-positive ECs between WT mice and rd1 mice, showing statistical differences at P5 and P8 (n = 6). d Statistical analysis of superficial vascular density showed a statistical difference during P3 to P8 between WT mice and rd1 mice (n = 6). e The number of tip cells was statistically different at P5 and P8 between WT mice and rd1 mice (n = 6). The data are presented as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. WT, wild type; IB4, isolectin B4; ECs, endothelial cells.

Close modal

The number of tip cells in the SVP was not significantly different between the WT and rd1 mice at P3 (Fig. 1b, d). The rd1 mice had fewer tip cells than WT mice at P5, while the rd1 mice had more tip cells than the WT mice at P8 (Fig. 1b, d).

Throughout SVP development, the retinal vessel density of the WT mice was always denser than that of the rd1 mice (Fig. 1b, e). At P11, there was no difference in the vessel density of SVP between the WT mice and the rd1 mice. However, this similarity disappeared at P13. During P15 to P21, the retinal SVP of the rd1 mice whose network remained relatively intact was slightly less dense than that of the WT mice (Fig. 2a, b).

Fig. 2.

SVP and deep vascular plexus (DVP) at P11 to P21. a Representative images of SVP and DVP of P11–P21 mice. Scale bar, 100 μm. b The density of SVP was statistically different between WT and rd1 mice during P15 to P21. c The density of DVP was statistically different between WT and rd1 mice during P11 to P21 (n = 4). The data are presented as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. WT, wild type; IB4, isolectin B4.

Fig. 2.

SVP and deep vascular plexus (DVP) at P11 to P21. a Representative images of SVP and DVP of P11–P21 mice. Scale bar, 100 μm. b The density of SVP was statistically different between WT and rd1 mice during P15 to P21. c The density of DVP was statistically different between WT and rd1 mice during P11 to P21 (n = 4). The data are presented as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. WT, wild type; IB4, isolectin B4.

Close modal

In both groups, the retinal deep vascular plexus (DVP) was not fully developed by P11, and there was a noticeable distance between the blood vessel network termini and the retinal periphery. Moreover, it was slightly less dense in the rd1 mice than in the WT mice. At P13, the vessel density of DVP in the WT mice was slightly but not significantly higher than that in the rd1 mice. In the rd1 mice, the retinal SVP and DVP rapidly degraded from P15 to P18, and the DVP was very sparse at P21. In contrast, the DVP in the WT mice remained stable from P13 to P21 (Fig. 2a, c). There was a significant difference in the vessel density of SVP and DVP between the WT and rd1 mice from P15 to P21 (Fig. 2b, c).

The Vascular Distributions in Different Retinal Layers of Rd1 Mice Lag behind Those of WT Mice

The retinal vascular distributions in the posterior pole, mid-peripheral, and peripheral parts of the retinal layers were compared using IB4 fluorescence staining of the retinal frozen sections at P8, P11, P13, P15, P18, and P21. At P8, the retinal blood vessels of the WT mice and rd1 mice were mainly localized at the superficial layer of the retinal posterior pole without significant difference. However, a few vessels started to develop into the retinal intermediate layers only in the WT mice. During P8 and P13, the blood vessels communicating between the superficial and intermediate layers were visible and showed no difference in the mice of both groups. In the rd1 mice, the retinal vessel density in the three parts of retina began to decrease from P15 onward, and the retinal SVP and DVP became very sparse at P21 (Fig. 3a–c). The peripheral retinal vasculatures of both groups were nearly equal only at P13. At all other timepoints, the retinal vessel density was higher in the WT mice than in the rd1 mice. By P15, the peripheral retinal vasculature reached stable in the WT mice but rapidly degraded in the rd1 mice (Fig. 3a, d).

Fig. 3.

Retinal vessel density in frozen sections from P8 to P21. a Retinal frozen sections were stained with IB4 (red) and DAPI (blue) to analyze the relative vessel density of different regions of the retina at different postnatal stages. Scale bar, 50 μm. b Quantification of the fluorescence intensity of intraretinal vessels in the posterior pole retina was found to be statistically different from P15 to P21 (n = 4). c Quantification of the fluorescence intensity of intraretinal vessels of the mid-peripheral retina was statistically different from P13 to P21 in each group (n = 4). d Quantification of the fluorescence intensity of intraretinal vessels of the peripheral retina was significantly different between P13 and P21 in each group (n = 4). The data are presented as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. WT, wild type; IB4, isolectin B4; DAPI, 4′,6-diamidino-2-phenylindole; GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; IPR, the inner part of the retina.

Fig. 3.

Retinal vessel density in frozen sections from P8 to P21. a Retinal frozen sections were stained with IB4 (red) and DAPI (blue) to analyze the relative vessel density of different regions of the retina at different postnatal stages. Scale bar, 50 μm. b Quantification of the fluorescence intensity of intraretinal vessels in the posterior pole retina was found to be statistically different from P15 to P21 (n = 4). c Quantification of the fluorescence intensity of intraretinal vessels of the mid-peripheral retina was statistically different from P13 to P21 in each group (n = 4). d Quantification of the fluorescence intensity of intraretinal vessels of the peripheral retina was significantly different between P13 and P21 in each group (n = 4). The data are presented as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. WT, wild type; IB4, isolectin B4; DAPI, 4′,6-diamidino-2-phenylindole; GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; IPR, the inner part of the retina.

Close modal

Changes of Retinal Thickness and RGC Degeneration Are Closely Related to Retinal Vessel Density in Rd1 Mice

Given that nearly 97% of photoreceptors are rods and localize in the ONL of the mouse retina, we evaluated the photoreceptors’ survival rate using the ONL thickness as an index [32, 33]. Examples of HE-stained sections of eye cups were shown in Figure 4a, and regions at 350 µm from the optic disk were enlarged and shown in Figure 4b. As the eye matures, the retinal thickness of WT mice exhibited a reduction in retinal thickness during P5–P8 and eventually stabilized at the end of the developmental period. Conversely, rd1 mice displayed a gradual attenuation and degeneration of retinal thickness from P5 to P21. The fluorescence intensity of RBPMS-positive RGCs in the rd1 mice gradually diminished from P11 onward (Fig. 4c). Here, the number of RGCs decreased in rd1 mice and there were significantly fewer RGCs in the rd1 mice than in the WT mice from P11 to P21 (Fig. 4c, i). This finding corresponded to the results of the HE staining (Fig. 4h).

Fig. 4.

Analysis of HE-stained retinal layer and retinal ganglion cells (RGCs). a Representative HE-stained image and 350 µm peri-optic nerve-labeled region. b Representative HE-stained images of two groups at different timepoints. Scale bar, 50 μm. c Representative images of retinal frozen sections stained with anti-RBPMS (green) and DAPI (blue). Scale bar, 50 μm. d Retinal index trends in two mouse groups (n = 3). e Positive correlation between rd1 mouse retinal vessel density and retinal thickness (r = 0.8883, p < 0.01). f Significant positive correlation between total vascular retinal density and IPR thickness of rd1 mice (r = 0.7172, p < 0.01). g Similar positive correlation with ONL thickness (r = 0.6893, p < 0.01). h, i, j Vascular densities (total, superficial, deep) correlate positively with RGC number (r = 0.8543, p < 0.01; r = 0.5255, p < 0.05; r = 0.6418, p < 0.01). k Anti-RBPMS antibody/DAPI (green/blue) staining showed statistically different RBPMS positive cells between WT and rd1 mice at P18 and P21 (n = 4). *p < 0.05, **p < 0.01, ***p < 0.001. WT, wild type; DAPI, 4′,6-diamidino-2-phenylindole; GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; IPR, the inner part of the retina.

Fig. 4.

Analysis of HE-stained retinal layer and retinal ganglion cells (RGCs). a Representative HE-stained image and 350 µm peri-optic nerve-labeled region. b Representative HE-stained images of two groups at different timepoints. Scale bar, 50 μm. c Representative images of retinal frozen sections stained with anti-RBPMS (green) and DAPI (blue). Scale bar, 50 μm. d Retinal index trends in two mouse groups (n = 3). e Positive correlation between rd1 mouse retinal vessel density and retinal thickness (r = 0.8883, p < 0.01). f Significant positive correlation between total vascular retinal density and IPR thickness of rd1 mice (r = 0.7172, p < 0.01). g Similar positive correlation with ONL thickness (r = 0.6893, p < 0.01). h, i, j Vascular densities (total, superficial, deep) correlate positively with RGC number (r = 0.8543, p < 0.01; r = 0.5255, p < 0.05; r = 0.6418, p < 0.01). k Anti-RBPMS antibody/DAPI (green/blue) staining showed statistically different RBPMS positive cells between WT and rd1 mice at P18 and P21 (n = 4). *p < 0.05, **p < 0.01, ***p < 0.001. WT, wild type; DAPI, 4′,6-diamidino-2-phenylindole; GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; IPR, the inner part of the retina.

Close modal

The retinal thickness and structure were comparably similar in both types of mice before P11. However, the IPR, the ONL, the full-retina thickness, and RGCs in rd1 mice underwent a precipitous degeneration since P15 (Fig. 4d). Total retinal vessel density (including superficial vascular density and deep vascular density) was closely and positively correlated with changes in total retinal thickness, the IPR thickness, and the ONL thickness (r = 0.8883, p < 0.01; r = 0.7036, p < 0.01; r = 0.6893, p < 0.01, respectively) (Fig. 4e–g). Changes in the density of total retinal vessels, SVP and DVP in rd1 mice were found to be positively correlated with the number of RGCs (r = 0.8543, p < 0.01; r = 0.5255, p < 0.05; r = 0.6418, p < 0.01, respectively) (Fig. 4h–J). RGCs underwent degeneration associated with retinal vascular degeneration (Fig. 4h–J).

Expressions of Vessel-Related CLD Are Decreased in Rd1 Mice

The expressions of four vessel-related CLD in rd1 mice and WT mice were investigated at different timepoints. The peak mRNA expression of CLD-1 and CLD-2 in rd1 mice was earlier than that of WT mice, but the overall mRNA expression levels of CLD-1 and CLD-2 were lower than those of WT mice. The mRNA expression levels of CLD-3 and CLD-5 in rd1 mice were lower than those of WT mice during the overall developmental period (Fig. 5a–d).

Fig. 5.

mRNA and protein expression of vessel-related claudin (CLD) in the retinas. a-d mRNA expression of CLD genes in each group (n = 3). e Western blot showing the expression of CLD1-3 and CLD-5 in the retinas of two group mice at different timepoints. f-i The histogram showed the densitometric analysis of the average levels of CLD proteins normalized to β-actin (n = 3). The data are expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. WT, wild type.

Fig. 5.

mRNA and protein expression of vessel-related claudin (CLD) in the retinas. a-d mRNA expression of CLD genes in each group (n = 3). e Western blot showing the expression of CLD1-3 and CLD-5 in the retinas of two group mice at different timepoints. f-i The histogram showed the densitometric analysis of the average levels of CLD proteins normalized to β-actin (n = 3). The data are expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. WT, wild type.

Close modal

During the retinal development period of WT mice, the protein expression of CLD-1 and CLD-5 showed a gradually increasing trend, while the protein expression of CLD-2 and CLD-3 showed a trend of increase at first and then decrease later (Fig. 5e). The protein expression levels of CLD-1, CLD-2, CLD-3, and CLD-5 in the retinas of rd1 mice were significantly lower than those of WT mice, especially at the late development stage when the degeneration of retinal deep blood vessels occurred (Fig. 5e, f). The protein expression of CLD-3 in the rd1 mice was extremely low at P21 (Fig. 5e, h).

Immunofluorescence staining was used to detect the expression of CLD-2 and CLD-3 in retinal frozen sections at P18, which is a period marked by the active formation of the BRB [34]. The co-localization between CLD-2 (green) and vascular marker, IB4 (red), was observed (Fig. 6a), indicating the expression of CLD-2 was in retinal vascular ECs. CLD-3 (green) was not co-stained with IB-4 (red) (Fig. 6a) but was found to co-localize in the NFL (Fig. 6d). These results indicated that CLD-3 was mainly expressed in the mouse RGCs, which was consistent with our previous research [21]. At P18, significant differences in vascular and neural-associated expressions were observed between the two groups. Compared to WT mice, the rd1 mice demonstrated a notable reduction in the expression level of CLD-2 and CLD-3, as well as in the RGCs and the retinal vessels (Fig. 6a–f).

Fig. 6.

Retinal immunofluorescence staining of CLD-2/CLD-3. a Representative images of retinal frozen sections stained with IB4 (red), anti-CLD-2 (green), anti-CLD-3 (green), and DAPI (blue) at P18. Scale bar, 20 μm. b, c There were statistical differences of fluorescence intensity of CLD-2/CLD-3 between WT and rd1 mice (n = 3). d Representative images of retinal frozen sections stained with anti-RBPMS (purple), anti-CLD-2 (green), anti-CLD-3 (green), and DAPI (blue) at P18. Scale bar, 20 μm. e, f There were statistical differences of fluorescence intensity between WT and rd1 mice (n = 3). The data are expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. WT, wild type; IB4, isolectin B4; DAPI, 4′,6-diamidino-2-phenylindole; GCL, ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer.

Fig. 6.

Retinal immunofluorescence staining of CLD-2/CLD-3. a Representative images of retinal frozen sections stained with IB4 (red), anti-CLD-2 (green), anti-CLD-3 (green), and DAPI (blue) at P18. Scale bar, 20 μm. b, c There were statistical differences of fluorescence intensity of CLD-2/CLD-3 between WT and rd1 mice (n = 3). d Representative images of retinal frozen sections stained with anti-RBPMS (purple), anti-CLD-2 (green), anti-CLD-3 (green), and DAPI (blue) at P18. Scale bar, 20 μm. e, f There were statistical differences of fluorescence intensity between WT and rd1 mice (n = 3). The data are expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001. WT, wild type; IB4, isolectin B4; DAPI, 4′,6-diamidino-2-phenylindole; GCL, ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer.

Close modal

Expressions of Angiogenesis-Related Proteins Decrease in Rd1 Mice

Vascular endothelial growth factor A (VEGFA) and vascular endothelial growth factor receptor 2 (VEGFR2) are closely associated with angiogenesis. Expression of VEGFA showed a gradually increasing trend in the WT mice but a gradually decreasing trend in the rd1 mice. And VEGFA was markedly lower in the rd1 mice than in the WT mice at all subsequent timepoints except at P3 (Fig. 7d, e). Expression of VEGFR2, which mediates VEGF signaling in ECs, was lower in rd1 mice than in WT mice (Fig. 7d, ) from P15 onward.

Fig. 7.

Protein expression of VEGFA/VEGFR2. a Western blot showed the expression of VEGFA and VEGFR2 in the retina of WT mice and rd1 mice at different timepoints. b, c The histogram showed the densitometric analysis of the average levels of VEGFA and VEGFR2 normalized to β-actin (n = 3). The data are expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. WT, wild type; VEGFA, vascular endothelial growth factor A; VEGFR2, vascular endothelial growth factor receptor 2.

Fig. 7.

Protein expression of VEGFA/VEGFR2. a Western blot showed the expression of VEGFA and VEGFR2 in the retina of WT mice and rd1 mice at different timepoints. b, c The histogram showed the densitometric analysis of the average levels of VEGFA and VEGFR2 normalized to β-actin (n = 3). The data are expressed as mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. WT, wild type; VEGFA, vascular endothelial growth factor A; VEGFR2, vascular endothelial growth factor receptor 2.

Close modal

Apoptosis detected by TUNEL staining was first observed in the ONL of rd1 mice at P8 (Fig. 8a). The most intensive apoptosis occurred in the retinas of rd1 mice at P11 and P15 due to the dramatic degeneration of the ONL, and the number of TUNEL-positive cells decreased at P18 and P21 (Fig. 8a, b). Interestingly, the TUNEL-positive cells were also found in the NFL at P11 (Fig. 8a). In contrast, fewer TUNEL-positive cells in the layers of the retina could be observed in WT mice at all timepoints (Fig. 8a).

Fig. 8.

TUNEL staining. a Representative images of retinal frozen sections stained with TUNEL of each group (n = 3). Scale bar, 50 μm. b Number of TUNEL-positive cells in rd1 mice retina. The data are presented as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. WT, wild type; GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; IPR, the inner part of the retina.

Fig. 8.

TUNEL staining. a Representative images of retinal frozen sections stained with TUNEL of each group (n = 3). Scale bar, 50 μm. b Number of TUNEL-positive cells in rd1 mice retina. The data are presented as the mean ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. WT, wild type; GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer; OPL, outer plexiform layer; ONL, outer nuclear layer; IPR, the inner part of the retina.

Close modal

Retinal vascular development in normal WT mice typically progresses through three stages [35]. Beginning on P1, vessels sprout from the optic disc head and spread outward. By P8, these superficial vessels reach the peripheral retina and then dive deeper to form the DVP. From P15, the superficial and deep retinal vascular networks start to form and fully establish by P21.

Our study indicated that by P8, retinal vessels in rd1 mice did not fully extend to the peripheral retina and this might result in more tip cells at the SVP in rd1 mice than WT mice. Fewer active vascular sprouts grow in rd1 mice than in WT mice, which might further affect the development of DVP. During P3–P13, the development of SVP and DVP of rd1 mice was slightly delayed but fully completed. However, from P15 onward, the retinal vessels in rd1 mice began to degenerate. By P21, the degeneration of retinal vasculature in rd1 mice was very noticeable. The SVP density of rd1 mice was significantly sparse compared to that of WT mice, and only a few residual branches and broken ends remained in DVP, which could not form a complete deep vascular network structure with normal function and shape. We also found retinal vessel degeneration in rd1 mice starting from small, deep retinal vessel branches in distal region to superficial large vessels in proximal area. This finding was consistent with previous studies [5, 36].

Angiogenesis involves novel blood vessel sprouting [37, 38]. It is commonly accepted that an abnormal hyperoxic state triggers retinal vascular regression. In other words, the characteristic retinal vascular attenuation is considered to be a secondary change occurring after photoreceptor loss [39]. The delayed development of SVP might be due to ocular blood supply insufficiency and mitochondrial stress in the early development period [40]. DVP network degeneration may be due to oxidative stress and blood oxygen supply-demand imbalances. This rd1 model aligns with peripheral-to-posterior pole vessel degeneration, which is observed in RP patients [1, 41]. The highest density of DVP in rd1 mice at P13 suggested the delayed deep vessel development, possibly due to the activation of full photoreceptor, accumulation of cyclic guanosine monophosphate, and reactions of oxidative stress [39, 42]. The vessel degeneration in our study was also consistent with previous studies [43, 44]. Oxidative stress disrupts the interaction between ECs and pericytes, which could lead to the responsiveness of ECs to VEGFA and hinder DVP formation [45‒48]. Therefore, retinal hyperoxygenation induced by an early loss of photoreceptors under the rd1 mutant background suppressed the expression of VEGFA/VEGFR2 and vessel-related CLD, including CLD-1, CLD-2, CLD-3, and CLD-5 in the retina of rd1 mice. CLD-5 is an important tight junction protein that constitutes the BRB, and the change of CLD-5 expression is related to VEGF [14, 48, 49]. The HIF2-alpha/VEGF feedback loop in Müller glial has been confirmed to be induced by severe hypoxia and acts on the growing retinal vasculature [48]. Additionally, there are interplays among CLD-1, CLD-2, CLD-3, and CLD-5 in the retina [14, 20, 50]. We can thus hypothesize that factors such as hyperoxia and decreased VEGFA in rd1 mice might act on retinal glial cells, ECs, and retinal neurons, thereby further affecting retinal neuro-vascular development and ultimately causing retinal neuro-vascular degeneration in rd1 mice. While some scholars have found that patients with RP have primary pathological reductions in retinal blood flow early in life. These manifestations are parts of the primary vascular dysregulation syndrome in RP, which leads to further photoreceptor damage and worsens the course of the disease [6, 51]. Therefore, vascular dysfunction in rd1 mice may be a result of metabolic changes but also possibly a primary alteration due to a genetic mutation. This inference still requires substantial follow-up studies.

We found that during developmental stages, changes in total retinal vascular density and apoptosis were positively correlated with the thickness of the ONL, which was consistent with previous study [5]. We hypothesized that in the developmental stage of rd1 mice, the choroidal circulation might be still unable to regulate apoptosis of the outer retina, retinal inner circulation, especially the DVP, but demonstrate a tolerance and regulatory response to the apoptosis of the photoreceptor cells. Previous studies have confirmed that DVP may satisfy the oxygen needs of photoreceptor cells’ inner segments [41]. Meanwhile, approximately 15% of the blood supply for photoreceptors near the macular fovea comes from the retinal circulation [52‒54]. This regulatory phenomenon of DVP in response to changes in oxygen supply is also observed in diabetic retinopathy and high myopia [55, 56]. The apoptosis of the outer retina occurring in the developmental stages of rd1 mice is sensitive to the oxygen supply from the DVP, but this does not contradict the fact that the main source of blood supply to the outer retinal layer primarily originates from the choroidal circulation [57]. The specific mechanism and the role of retinal blood supply in neurovascular degeneration in rd1 mice is worth an in-depth study.

RGCs are unique among retinal neurons as they directly project their axons into the central nervous system and serve a critical role in visual function [58]. Loss of RGC in rd1 mice at 1 year of age does not exceed 6% of those in WT mice [59]. However, Steven et al. [60] have also found that rd1 mice develop a decreased ganglion cell response later in development. In this study, we proved the loss and degeneration of RGCs was at the late stage of the developmental period in rd1 mice, and the expression of CLD-3 was a significant reduction, which was closely related to RGCs [20, 21]. These findings are consistent with previous studies using disease models of RGC injury [20]. Programmed cell death of RGCs in WT mice begins at P2, continues throughout development, and ends in adulthood [61]. We found that the loss of RGCs was related to changes in SVP and DVP and the degeneration of retinal vessels was earlier than the loss of RGCs in rd1 mice. RGCs derive their blood supply from all layers of the retinal vasculature and their axons run through the retina. RGCs have certain physiological remodeling and compensatory functions, so a therapeutic time window would occur between vascular degeneration and neuro-degeneration in rd1 mice [62]. We proposed that interventions targeting retinal vasculature might bear greater practical significance in the early treatment of retinal degenerative diseases. Our previous research has demonstrated that downregulation of CLD-3 can induce apoptosis of RGCs by suppressing the expression of Bcl-2 and VEGFA [21, 22]. CLD-3 functions as an intermediate junction station for proteins and pathways associated with retinal nerve and vascular development [50, 63]. Our previous study has indicated that the downregulation of CLD-3 in zebrafish might have an impact on the Notch pathway [22]. In this study, we have observed a reduction in CLD-3 levels, concomitant with the downregulation of VEGFA/VEGFR2 in rd1 mice. Therefore, loss of RGC might be due to hypoxia, retinal vessel degeneration, or downregulation of CLD-3 [20, 64]. These needed further in-depth investigations.

This study demonstrated that rd1 mouse had characteristics of retinal vasculature degeneration and retinal neuro-degeneration of ONL and RGCs. rd1 mouse also showed a notable decline in the expression of vessel-related CLD proteins, which might play a pivotal role in neurovascular development. The data indicated rd1 mouse is a valuable animal model of not only retinal neuro-degeneration but also retinal vascular degeneration and remodeling.

This study protocol was reviewed and approved by the Institutional Review Board of Zhongshan Ophthalmic Center (Guangzhou, China), Approval No. Z2021080.

The authors have no conflicts of interest to declare.

This work was supported by the National Natural Science Foundation of China to Yan Luo (81770971, 81371020).

Aoxiang Wang completed experiments and wrote paper. Jinxi Zhou completed image processing. Yiwen Hong, Yishen Wang, and Jianying Pan supported the data analysis. Yamei Cui and Yue Wu reviewed the paper and gave suggestions on it. Yan Luo contributed to the experiment design and paper supervision.

Data are not available due to ethical reasons. Further inquiries can be directed to the corresponding author.

1.
Kalloniatis
M
,
Nivison-Smith
L
,
Chua
J
,
Acosta
ML
,
Fletcher
EL
.
Using the rd1 mouse to understand functional and anatomical retinal remodelling and treatment implications in retinitis pigmentosa: a review
.
Exp Eye Res
.
2016
;
150
:
106
21
.
2.
Pagon
RA
.
Retinitis pigmentosa
.
Surv Ophthalmol
.
1988
;
33
(
3
):
137
77
.
3.
Verbakel
SK
,
van Huet
RAC
,
Boon
CJF
,
den Hollander
AI
,
Collin
RWJ
,
Klaver
CCW
, et al
.
Non-syndromic retinitis pigmentosa
.
Prog Retin Eye Res
.
2018
;
66
:
157
86
.
4.
Liu
R
,
Lu
J
,
Liu
Q
,
Wang
Y
,
Cao
D
,
Wang
J
, et al
.
Effect of choroidal vessel density on the ellipsoid zone and visual function in retinitis pigmentosa using optical coherence tomography angiography
.
Invest Ophthalmol Vis Sci
.
2019
;
60
(
13
):
4328
35
.
5.
Ripolles-Garcia
A
,
Chen
Y
,
Sato
Y
,
Gray
A
,
Ying
GS
,
Aguirre
GD
, et al
.
Retinal vascular plexuses are unequally affected in canine inherited retinal degenerations
.
Invest Ophthalmol Vis Sci
.
2022
;
63
(
12
):
22
.
6.
Wolf
S
,
Pöstgens
H
,
Bertram
B
,
Schulte
K
,
Teping
C
,
Reim
M
.
[Hemodynamic findings in patients with retinitis pigmentosa]
.
Klin Monbl Augenheilkd
.
1991
;
199
(
11
):
325
9
.
7.
Jung
YH
,
Kwak
JJ
,
Joo
K
,
Lee
HJ
,
Park
KH
,
Kim
MS
, et al
.
Clinical and genetic features of Koreans with retinitis pigmentosa associated with mutations in rhodopsin
.
Front Genet
.
2023
;
14
:
1240067
.
8.
Daich Varela
M
,
Esener
B
,
Hashem
SA
,
Cabral de Guimaraes
TA
,
Georgiou
M
,
Michaelides
M
.
Structural evaluation in inherited retinal diseases
.
Br J Ophthalmol
.
2021
;
105
(
12
):
1623
31
.
9.
Rizzolo
LJ
,
Peng
S
,
Luo
Y
,
Xiao
W
.
Integration of tight junctions and claudins with the barrier functions of the retinal pigment epithelium
.
Prog Retin Eye Res
.
2011
;
30
(
5
):
296
323
.
10.
Turksen
K
,
Troy
TC
.
Barriers built on claudins
.
J Cell Sci
.
2004
;
117
(
Pt 12
):
2435
47
.
11.
Singh
AB
,
Dhawan
P
.
Claudins and cancer: fall of the soldiers entrusted to protect the gate and keep the barrier intact
.
Semin Cell Dev Biol
.
2015
;
42
:
58
65
.
12.
Hashimoto
Y
,
Okada
Y
,
Shirakura
K
,
Tachibana
K
,
Sawada
M
,
Yagi
K
, et al
.
Anti-claudin antibodies as a concept for development of claudin-directed drugs
.
J Pharmacol Exp Ther
.
2019
;
368
(
2
):
179
86
.
13.
Luo
Y
,
Xiao
W
,
Zhu
X
,
Mao
Y
,
Liu
X
,
Chen
X
, et al
.
Differential expression of claudins in retinas during normal development and the angiogenesis of oxygen-induced retinopathy
.
Invest Ophthalmol Vis Sci
.
2011
;
52
(
10
):
7556
64
.
14.
Tian
R
,
Luo
Y
,
Liu
Q
,
Cai
M
,
Li
J
,
Sun
W
, et al
.
The effect of claudin-5 overexpression on the interactions of claudin-1 and -2 and barrier function in retinal cells
.
Curr Mol Med
.
2014
;
14
(
9
):
1226
37
.
15.
Fang
M
,
Wan
W
,
Li
Q
,
Wan
W
,
Long
Y
,
Liu
H
, et al
.
Asiatic acid attenuates diabetic retinopathy through TLR4/MyD88/NF-κB p65 mediated modulation of microglia polarization
.
Life Sci
.
2021
;
277
:
119567
.
16.
Pope
JL
,
Bhat
AA
,
Sharma
A
,
Ahmad
R
,
Krishnan
M
,
Washington
MK
, et al
.
Claudin-1 regulates intestinal epithelial homeostasis through the modulation of Notch-signalling
.
Gut
.
2014
;
63
(
4
):
622
34
.
17.
Tabariès
S
,
Annis
MG
,
Lazaris
A
,
Petrillo
SK
,
Huxham
J
,
Abdellatif
A
, et al
.
Claudin-2 promotes colorectal cancer liver metastasis and is a biomarker of the replacement type growth pattern
.
Commun Biol
.
2021
;
4
(
1
):
657
.
18.
Kakogiannos
N
,
Ferrari
L
,
Giampietro
C
,
Scalise
AA
,
Maderna
C
,
Ravà
M
, et al
.
JAM-A acts via C/EBP-α to promote claudin-5 expression and enhance endothelial barrier function
.
Circ Res
.
2020
;
127
(
8
):
1056
73
.
19.
Daugherty
BL
,
Ward
C
,
Smith
T
,
Ritzenthaler
JD
,
Koval
M
.
Regulation of heterotypic claudin compatibility
.
J Biol Chem
.
2007
;
282
(
41
):
30005
13
.
20.
Zhao
Z
,
Yu
X
,
Yang
X
,
Zhang
J
,
Zhang
D
,
Sun
N
, et al
.
Elevated intraocular pressure causes cellular and molecular retinal injuries, advocating a more moderate intraocular pressure setting during phacoemulsification surgery
.
Int Ophthalmol
.
2020
;
40
(
12
):
3323
36
.
21.
Cao
D
,
Li
J
,
Wang
X
,
Wang
J
,
Liu
R
,
Lu
J
, et al
.
The effect of AAV-mediated downregulation of Claudin-3 on the development of mouse retinal vasculature
.
Exp Eye Res
.
2021
;
213
:
108836
.
22.
Lu
J
,
Liu
R
,
Miao
A
,
Chen
X
,
Xiao
W
,
Wang
Y
, et al
.
The role of cldnh during the early retinal development in zebrafish
.
Exp Eye Res
.
2020
;
200
:
108207
.
23.
Yan
W
,
Yao
L
,
Liu
W
,
Sun
K
,
Zhang
Z
,
Zhang
L
.
A kind of rd1 mouse in C57BL/6J mice from crossing with a mutated Kunming mouse
.
Gene
.
2017
;
607
:
9
15
.
24.
Keeler
C
.
Retinal degeneration in the mouse is rodless retina
.
J Hered
.
1966
;
57
(
2
):
47
50
.
25.
Mahato
B
,
Kaya
KD
,
Fan
Y
,
Sumien
N
,
Shetty
RA
,
Zhang
W
, et al
.
Pharmacologic fibroblast reprogramming into photoreceptors restores vision
.
Nature
.
2020
;
581
(
7806
):
83
8
.
26.
Cui
Y
,
Liu
R
,
Hong
Y
,
Wang
Y
,
Zhu
Y
,
Wen
T
, et al
.
MicroRNA-92a-3p regulates retinal angiogenesis by targeting SGK3 in vascular endothelial cells
.
Invest Ophthalmol Vis Sci
.
2022
;
63
(
11
):
19
.
27.
Hong
Y
,
Wang
Y
,
Cui
Y
,
Pan
J
,
Mao
S
,
Zhu
Y
, et al
.
MicroRNA-124-3p attenuated retinal neovascularization in oxygen-induced retinopathy mice by inhibiting the dysfunction of retinal neuroglial cells through STAT3 pathway
.
Int J Mol Sci
.
2023
;
24
,
11767
(
14
).
28.
Wang
Y
,
Hong
Y
,
Mao
S
,
Jiang
Y
,
Cui
Y
,
Pan
J
, et al
.
An interaction-based method for refining results from gene set enrichment analysis
.
Front Genet
.
2022
;
13
:
890672
.
29.
Carroll
LS
,
Uehara
H
,
Fang
D
,
Choi
S
,
Zhang
X
,
Singh
M
, et al
.
Intravitreal aav2.comp-ang1 attenuates deep capillary plexus expansion in the aged diabetic mouse retina
.
Invest Ophthalmol Vis Sci
.
2019
;
60
(
7
):
2494
502
.
30.
Rösch
S
,
Aretzweiler
C
,
Müller
F
,
Walter
P
.
Evaluation of retinal function and morphology of the pink-eyed royal college of surgeons (RCS) rat: a comparative study of in vivo and in vitro methods
.
Curr Eye Res
.
2017
;
42
(
2
):
273
81
.
31.
Zantut
PRA
,
Veras
MM
,
Benevenutto
SGM
,
Safatle
AMV
,
Pecora
RA
,
Yariwake
VY
, et al
.
Lasting effects of prenatal exposure to Cannabis in the retina of the offspring: an experimental study in mice
.
Int J Retina Vitreous
.
2021
;
7
(
1
):
45
.
32.
Song
S
,
Bao
S
,
Zhang
C
,
Zhang
J
,
Lv
J
,
Li
X
, et al
.
Stimulation of AMPK prevents diabetes-induced photoreceptor cell degeneration
.
Oxid Med Cel Longev
.
2021
;
2021
:
5587340
.
33.
A
L
,
Zou
T
,
He
J
,
Chen
X
,
Sun
D
,
Fan
X
, et al
.
Rescue of retinal degeneration in rd1 mice by intravitreally injected metformin
.
Front Mol Neurosci
.
2019
;
12
:
102
.
34.
Tawfik
A
,
Samra
YA
,
Elsherbiny
NM
,
Al-Shabrawey
M
.
Implication of hyperhomocysteinemia in blood retinal barrier (BRB) dysfunction
.
Biomolecules
.
2020
;
10
,
1119
(
8
).
35.
Mazzoni
J
,
Smith
JR
,
Shahriar
S
,
Cutforth
T
,
Ceja
B
,
Agalliu
D
.
The wnt inhibitor Apcdd1 coordinates vascular remodeling and barrier maturation of retinal blood vessels
.
Neuron
.
2017
;
96
(
5
):
1055
69.e6
.
36.
Hanna
J
,
Yücel
YH
,
Zhou
X
,
Mathieu
E
,
Paczka-Giorgi
LA
,
Gupta
N
.
Progressive loss of retinal blood vessels in a live model of retinitis pigmentosa
.
Can J Ophthalmol
.
2018
;
53
(
4
):
391
401
.
37.
Nitzsche
B
,
Rong
WW
,
Goede
A
,
Hoffmann
B
,
Scarpa
F
,
Kuebler
WM
, et al
.
Coalescent angiogenesis-evidence for a novel concept of vascular network maturation
.
Angiogenesis
.
2022
;
25
(
1
):
35
45
.
38.
Risau
W
.
Mechanisms of angiogenesis
.
Nature
.
1997
;
386
(
6626
):
671
4
.
39.
Lau
JC
,
Linsenmeier
RA
.
Oxygen consumption and distribution in the Long-Evans rat retina
.
Exp Eye Res
.
2012
;
102
:
50
8
.
40.
Jiang
K
,
Mondal
AK
,
Adlakha
YK
,
Gumerson
J
,
Aponte
A
,
Gieser
L
, et al
.
Multiomics analyses reveal early metabolic imbalance and mitochondrial stress in neonatal photoreceptors leading to cell death in Pde6brd1/rd1 mouse model of retinal degeneration
.
Hum Mol Genet
.
2022
;
31
(
13
):
2137
54
.
41.
Rahimi
M
,
Leahy
S
,
Matei
N
,
Blair
NP
,
Jeong
S
,
Craft
CM
, et al
.
Assessment of inner retinal oxygen metrics and thickness in a mouse model of inherited retinal degeneration
.
Exp Eye Res
.
2021
;
205
:
108480
.
42.
Tiriac
A
,
Smith
BE
,
Feller
MB
.
Light prior to eye opening promotes retinal waves and eye-specific segregation
.
Neuron
.
2018
;
100
(
5
):
1059
65.e4
.
43.
Pennesi
ME
,
Nishikawa
S
,
Matthes
MT
,
Yasumura
D
,
LaVail
MM
.
The relationship of photoreceptor degeneration to retinal vascular development and loss in mutant rhodopsin transgenic and RCS rats
.
Exp Eye Res
.
2008
;
87
(
6
):
561
70
.
44.
Nguyen
M
,
Sullivan
J
,
Shen
W
.
Retinal vascular remodeling in photoreceptor degenerative disease
.
Exp Eye Res
.
2023
;
234
:
109566
.
45.
Su
L
,
Gong
X
,
Fan
R
,
Ni
T
,
Yang
F
,
Zhang
X
, et al
.
Mechanism of action of platinum nanoparticles implying from antioxidant to metabolic programming in light-induced retinal degeneration model
.
Redox Biol
.
2023
;
65
:
102836
.
46.
Huang
H
.
Pericyte-endothelial interactions in the retinal microvasculature
.
Int J Mol Sci
.
2020
;
21
:
7413
(
19
).
47.
Fukuda
S
,
Ohneda
O
,
Oshika
T
.
Oxidative stress retards vascular development before neural degeneration occurs in retinal degeneration rd1 mice
.
Graefes Arch Clin Exp Ophthalmol
.
2014
;
252
(
3
):
411
6
.
48.
Rattner
A
,
Wang
Y
,
Zhou
Y
,
Williams
J
,
Nathans
J
.
The role of the hypoxia response in shaping retinal vascular development in the absence of Norrin/Frizzled4 signaling
.
Invest Ophthalmol Vis Sci
.
2014
;
55
(
12
):
8614
25
.
49.
Bora
K
,
Kushwah
N
,
Maurya
M
,
Pavlovich
MC
,
Wang
Z
,
Chen
J
.
Assessment of inner blood-retinal barrier: animal models and methods
.
Cells
.
2023
;
12
,
2443
(
20
).
50.
Liu
F
,
Xu
T
,
Peng
S
,
Adelman
RA
,
Rizzolo
LJ
.
Claudins regulate gene and protein expression of the retinal pigment epithelium independent of their association with tight junctions
.
Exp Eye Res
.
2020
;
198
:
108157
.
51.
Konieczka
K
,
Flammer
AJ
,
Todorova
M
,
Meyer
P
,
Flammer
J
.
Retinitis pigmentosa and ocular blood flow
.
EPMA J
.
2012
;
3
(
1
):
17
.
52.
Birol
G
,
Wang
S
,
Budzynski
E
,
Wangsa-Wirawan
ND
,
Linsenmeier
RA
.
Oxygen distribution and consumption in the macaque retina
.
Am J Physiol Heart Circ Physiol
.
2007
;
293
(
3
):
H1696
704
.
53.
Snodderly
DM
,
Weinhaus
RS
,
Choi
JC
.
Neural-vascular relationships in central retina of macaque monkeys (Macaca fascicularis)
.
J Neurosci
.
1992
;
12
(
4
):
1169
93
.
54.
Linsenmeier
RA
,
Zhang
HF
.
Retinal oxygen: from animals to humans
.
Prog Retin Eye Res
.
2017
;
58
:
115
51
.
55.
Wu
Q
,
Chen
Q
,
Lin
B
,
Huang
S
,
Wang
Y
,
Zhang
L
, et al
.
Relationships among retinal/choroidal thickness, retinal microvascular network and visual field in high myopia
.
Acta Ophthalmol
.
2020
;
98
(
6
):
e709
14
.
56.
Yi
J
,
Liu
W
,
Chen
S
,
Backman
V
,
Sheibani
N
,
Sorenson
CM
, et al
.
Visible light optical coherence tomography measures retinal oxygen metabolic response to systemic oxygenation
.
Light Sci Appl
.
2015
;
4
(
9
):
e334
.
57.
Linsenmeier
RA
,
Dmitriev
AV
,
Dmitriev
AA
.
Oxygen profiles and oxygen consumption in the isolated mouse retina
.
Exp Eye Res
.
2023
;
233
:
109554
.
58.
Yao
F
,
Peng
J
,
Zhang
E
,
Ji
D
,
Gao
Z
,
Tang
Y
, et al
.
Pathologically high intraocular pressure disturbs normal iron homeostasis and leads to retinal ganglion cell ferroptosis in glaucoma
.
Cell Death Differ
.
2023
;
30
(
1
):
69
81
.
59.
Damiani
D
,
Novelli
E
,
Mazzoni
F
,
Strettoi
E
.
Undersized dendritic arborizations in retinal ganglion cells of the rd1 mutant mouse: a paradigm of early onset photoreceptor degeneration
.
J Comp Neurol
.
2012
;
520
(
7
):
1406
23
.
60.
Stasheff
SF
.
Emergence of sustained spontaneous hyperactivity and temporary preservation of OFF responses in ganglion cells of the retinal degeneration (rd1) mouse
.
J Neurophysiol
.
2008
;
99
(
3
):
1408
21
.
61.
Voyatzis
S
,
Muzerelle
A
,
Gaspar
P
,
Nicol
X
.
Modeling activity and target-dependent developmental cell death of mouse retinal ganglion cells ex vivo
.
PLoS One
.
2012
;
7
(
2
):
e31105
.
62.
Telias
M
,
Sit
KK
,
Frozenfar
D
,
Smith
B
,
Misra
A
,
Goard
MJ
, et al
.
Retinoic acid inhibitors mitigate vision loss in a mouse model of retinal degeneration
.
Sci Adv
.
2022
;
8
(
11
):
eabm4643
.
63.
Reinhold
AK
,
Rittner
HL
.
Barrier function in the peripheral and central nervous system-a review
.
Pflugers Arch
.
2017
;
469
(
1
):
123
34
.
64.
Brockhaus
K
,
Melkonyan
H
,
Prokosch-Willing
V
,
Liu
H
,
Thanos
S
.
Alterations in tight- and adherens-junction proteins related to glaucoma mimicked in the organotypically cultivated mouse retina under elevated pressure
.
Invest Ophthalmol Vis Sci
.
2020
;
61
(
3
):
46
.