The blood-retinal barrier (BRB) is a biological unit comprised of specialized capillary endothelial cells firmly connected by intercellular tight junctions and endothelium-surrounding glial cells. The BRB is essential for maintaining the retinal microenvironment and low permeability and is compromised in an early phase during the progression of diabetic retinopathy. Here, we demonstrate that retinoic acid receptor (RAR)α stimulants preferentially act on glial cells rather than endothelial cells, resulting in the enhanced expression of glial cell line–derived neurotrophic factor (GDNF) through recruitment of the RARα-driven trans-acting coactivator to the 5′-flanking region of the gene promoter. Conversely, RARα decreases expression of vascular endothelial growth factor (VEGF)/vascular permeability factor. These gene expression alterations causally limit vascular permeability by modulating the tight junction function of capillary endothelium in a paracrine manner in vitro. The phenotypic transformation of glial cells mediated by RARα is sufficient for significant reductions of vascular leakage in the diabetic retina, suggesting that RARα antagonizes the loss of tight junction integrity induced by diabetes. These findings reveal that glial cell–derived cytokines such as GDNF and VEGF regulate BRB function, implying that the glial cell can be a possible therapeutic target in diabetic retinopathy.
The blood-retinal barrier (BRB) is a biological unit of retinal vessels with a well-differentiated network, including glial cells such as astrocytes and Müller cells, maintaining the retinal microenvironment and low permeability. The substantial apparatus of the BRB is a barrier comprised of tight junctions between the capillary endothelial cells that strictly regulate the paracellular pathways between the cells (1). BRB breakdown is closely associated with a number of retinal diseases such as diabetic retinopathy, which is characterized by vascular leakage due to increased vascular permeability in its early pathogenesis (2).
A number of studies have demonstrated that hyperglycemia in diabetes increases the expression of vascular endothelial growth factor (VEGF)/vascular permeability factor (3,4), which is believed to be a critical factor in the development of diabetic retinopathy (5–7). However, the molecular pharmacology that directly inhibits activated VEGF has not been proven to satisfactorily block microangiopathy in diabetic retinopathy (8,9). While glial cell line–derived neurotrophic factor (GDNF) was originally identified as a neurotrophic differentiation factor for dopaminergic neurons in the central nervous system and retina (10,11), we have proposed the novel idea that GDNF secreted from glial cells is a critical factor in regulating the vascular permeability of the BRB in a biological unit comprised of capillary endothelial cells and glial cells via modulation of the barrier function of tight junctions (12–14). In addition, we have also demonstrated that certain advanced glycation end products that are formed under hyperglycemic conditions increase the vascular permeability of the BRB in vitro, which is achieved by the induction of VEGF and reduction of GDNF expression from glial cells, suggesting that phenotypic alteration of glial cells in diabetes is responsible for the BRB breakdown (12–14).
The vitamin A metabolite all-trans retinoic acid (ATRA) is a potent regulator of cell differentiation and an essential signaling molecule in embryonic development and throughout life (15,16). A previous study has shown that ATRA can differentiate pluripotent embryonal carcinoma cells into neuronal and glial tissues and that it plays an important role in the induction of GDNF responsiveness in these cells (17). This evidence prompted us to investigate whether retinoic acids such as ATRA act on glial cells and, consequently, whether they have any effects on endothelial cells in the BRB. Here, we show that retinoic acid receptor (RAR)α stimulation preferentially alters the gene expression of glial cell–derived cytokines such as GDNF and VEGF, which are responsible for regulating BRB integrity, resulting in the modulation of tight junction functions of BRB-forming capillary endothelium. Our findings imply that the glial cell can be a likely candidate for a therapeutic target in diabetic retinopathy.
RESEARCH DESIGN AND METHODS
Cell culture and treatment.
The human glioblastoma cell line U373MG and endothelial cells cultured using the bovine brain microvascular endothelial cell system (bMVEC-B) (Cambrex, Baltimore, MD) were maintained in Dulbecco's modified Eagle's medium (Sigma, St. Louis, MO) supplemented with 10% fetal bovine serum (FBS) (Cell Culture Technologies, Lugano, Switzerland) or charcoal dextran–treated FBS (18), with 100 units/ml penicillin, and with 100 μg/ml streptomycin (Sigma). Primary culture of human astrocytes (Cambrex) was maintained in astrocyte growth medium (Cambrex). The 8th to 12th passages of bMVEC-B and 3rd to 4th passages of human astrocytes were used in this study. U373MG cells and human astrocytes were treated within a dose ranging from 10 nmol/l to 1 μmol/l ATRA or with 100 nmol/l ATRA, 100 nmol/l 9-cis retinoic acid (9cRA), and 10 nmol/l 4-[(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl)carboxamido] benzoic acid (Am580). Confluent cultured endothelial cells were treated with 1 ng/ml GDNF recombinant protein (GDNFr-p) (PeproTech, Rocky Hill, NJ) or 100 nmol/l ATRA for up to 7 days, and RNA and protein samples were extracted from the cells treated for 7 days. For transfection of short interfering RNA (siRNA), glial cells were transfected with 150 nmol/l GDNF siRNA (Santa Cruz Biotechnologies, Santa Cruz, CA) by electroporation (capacitance extender 2.0 pulse/960 μPD; Bio-Rad, Hercules, CA). Unless otherwise specified, cells treated for 4 and 8 h were used for RNA and protein preparation, respectively.
Real-time PCR and RT-PCR analysis.
Total RNA was extracted from cells using TRIzol Reagent (Invitrogen, Carisbad, CA). Then, RNA (1 μg) was reverse transcribed using poly-T oligonucleotide and M-MuLV reverse transcriptase (Invitrogen). For analysis of gene expression, the genes of interest were amplified from dilutions of cDNA with the TaqMan gene expression assay using ABI Prism 7000 (Applied Biosystems, Foster City, CA) or of PCR using specific sense and antisense primers for up to 40 cycles. We examined various cycling parameters for each PCR experiment to define optimal conditions for linearity to allow for semiquantitative analysis of signal intensity. To provide a qualitative control for reaction efficiency, PCRs were performed with primers coding for glyceraldehyde-3-phosphate dehydrogenase (GAPDH). (All primer sequences used in this study are available from the authors on request.) For the densitometric analysis, signals in RT-PCR analysis were quantitated using Scion Image 1.62 (Scion Corporation, Frederick, MD).
Assessment of endothelial permeability.
Endothelial cells were grown to confluence on transwell semipermeable membranes (pore size 0.4 μm; Costar, Cambridge, MA) in the presence or absence of 100 nmol/l ATRA or 1 ng/ml GDNFr-p in the lower chamber. In the coculture experiments, glial cells cultured in the lower chamber were treated with 100 nmol/l ATRA or 10 nmol/l Am580 for 8 h and cocultured with endothelial cells that were grown to confluence on transwell membranes in the upper chamber. Paracellular tracer flux was measured by applying [14C]inulin (MW 5 kDa) at 5 × 105 dpm/well onto an endothelial monolayer in the apical compartment, and the samples were collected from the basolateral compartment in a time-dependent manner. Radioactivity of [14C] was counted using a scintillation counter (Beckman LS6500; Beckman Coulter, Fullerton, CA).
Immunohistochemistry.
Confluent-cultured endothelial cells treated with GDNFr-p were immunolabeled with antibodies directed against claudin-5, occludin, and zonula occludens-1 (all from Invitrogen). Samples were then probed with corresponding secondary antibodies coupled with Alexa Fluor 594 (Invitrogen), and cells were examined using a laser-scanning confocal microscope (MRC 1024; Bio-Rad).
Quantification of GDNF and VEGF proteins.
GDNF and VEGF proteins were quantitated by enzyme-linked immunosorbent assay (ELISA) using a GDNF Emax Immunoassay system (Promega, Madison, WI) or VEGF Immunoassay kit (Biosource, Camarillo, CA). In addition, GDNF and VEGF proteins were also examined by Western blot analysis. The protein concentration was determined with a bicinchoninic acid protein assay kit (Pierce, Rockford, IL). Twenty micrograms of each denatured protein was subjected to SDS-PAGE. The membrane was reacted with antibodies against GDNF, VEGF, and β-actin (all from Santa Cruz), and the immunoreactions were visualized using an enhanced chemiluminescent system (Amersham Bioscience, Buckinghamshire, U.K.).
Human vitreous samples.
Undiluted vitreous samples were obtained from 85 consecutive patients with diabetic retinopathy (65 patients) and macular holes (20 nondiabetic control subjects), who underwent vitrectomy (supplementary Table [available in an online appendix at http://dx.doi.org/10.2337/db06-1431]). Before the experiment, informed consent was obtained from each patient, and the experiment was approved by the institutional review board.
GDNF promoter assay.
We found a putative human GDNF promoter fragment in the gene database (http://www.ncbi.nlm.nih.gov) based on the exon 1 sequence of GDNF cDNA (GeneBank accession no. NM_199234) and amplified it by genomic PCR using a downstream primer encompassing the transcription initiation codon 5′-AGCCACGACATCCCATAACT-3′ and forward primer 5′-TGCACTTTTCCTGTCTGC-3′, which corresponded to the sequence 1,793 bp (∼1.8 kb) upstream from the transcriptional initiation site. Since three p300 binding motifs were found in this region using a Web-based search for putative nuclear receptors responsible for GDNF expression (http://motif.genome.jp), we also amplified a 1.2-kb fragment that lacked all p300 binding sites using forward primer 5′-GGCACTGAGGTTTTTGCATT-3′ and reverse primer 5′-CTTCTCCACCACACGGTCTT-3′. After performing TA cloning of 1.8 kb and 1.2 kb GDNF promoter cDNAs using pCR 2.1 (Invitrogen), we cloned each cDNA into pGL-3Basic vector (Promega). HeLa cells (1 × 105) were split for seeding into 24-well plates 24 h before transfection and transfected to each vector (4 μg) with 40 ng pRL-Tk (Promega) in 4 μl FuGENE 6 reagent (Roche Diagnostics, Basel, Switzerland). After 4 h treatment with 100 nmol/l ATRA or 10 nmol/l Am580, proteins were extracted using a passive lysis buffer (Promega) and were read using dual luciferase reagents (Promega) and an AB-2200 luminometer (Atto, Tokyo, Japan).
Glial cells treated with 100 nmol/l ATRA or 10 nmol/l Am580 for 4 h were subjected to chromatin immunoprecipitation (ChIP) assay. Aliquots from the cells were incubated with specific antibodies against p300 (Santa Cruz) and rotated overnight at 4°C. The immunocomplex was recovered by centrifugation and DNA extracted. DNA from the bulky lysate of the cells was directly amplified as a positive control, and a DNA template was isolated from the lysate without the immunoprecipitation step as a negative control.
Animal experiments.
C57BL/6 male mice (5 weeks old; CLEA, Tokyo, Japan) were housed in a specific pathogen-free facility and were maintained on standard mouse diet and water. To induce diabetes, mice were intraperitoneally injected with 40 mg/kg streptozotocin (Sigma) for 5 consecutive days. Control mice were intraperitoneally injected with a solvent alone (250 μl DMSO; Sigma). Six weeks after the verification of diabetes, mice were treated with 1.0 mg/kg ATRA every day, 3.75 mg/kg Am580 every other day, or DMSO every day for 1 week. To examine the leakage of retinal vessels, we injected 50 mg/kg fluorescein isothiocyanate (FITC)-dextran (4 kDa; Sigma) into mice via the vena cava, following which the mice were killed, and the bilateral eyes were enucleated 5 min after the FITC injection under general anesthesia. The aqueous phase of the sonicated right eye was examined to determine the FITC concentration (WALLAC 1420; Perkin Elmer, Wellesley, MA) and for quantification of GDNF and VEGF proteins. Left eyes were flat mounted, and the retinas were analyzed by laser-scanning confocal analysis. The FITC concentration in cardiac blood of each mouse was calculated to provide a quantitative control. All animal experiments were performed in accordance with the Guidelines for Animal Experiments of Sapporo Medical University School of Medicine.
Statistical analysis.
All values are expressed as means and SDs of at least three independent experiments. Data were statistically analyzed with the Mann-Whitney U test for comparison of two groups and Fisher's test for comparison of three or more groups. Statistical significance was set at P < 0.05.
RESULTS
RARα stimulants upregulate GDNF expression in glial cells.
ATRA is a biologically active regulator for cell fate decision, having a broad range of functions involving cell differentiation, proliferation, and apoptosis in various cell types (15,16). Based on the recent observation that ATRA plays an important role in the induction of GDNF expression and responsiveness of glial cells (17), we examined whether ATRA could regulate the competence of glial cells to affect GDNF expression. We first found that ATRA upregulated GDNF mRNA expression in a time- and dose-dependent manner in U373MG cells (Fig. 1A and B), which express glial fibrillary acidic protein (GFAP), a specific differentiation marker of glial cells. ATRA clearly upregulated GDNF mRNA in a dose ranging from 10 nmol/l to 1 μmol/l ATRA, and the peak increase in the level of GDNF mRNA was achieved at 100 nmol/l (Fig. 1A and B). The induced level of GDNF mRNA was almost the same between 3 and 24 h after the treatment, indicating that U373MG required 3 h for full induction of GDNF mRNA (Fig. 1A). While GDNF expression was not changed in the medium supplemented with endogenous retinoic acid–depleted (i.e., charcoal-dextran treated) FBS, treatment with 100 nmol/l ATRA resulted in enhanced expression of GDNF (Fig. 1C), suggesting that the upregulation of GDNF required pharmacological doses of ATRA.
Our previous studies have demonstrated that GDNF is an important factor in regulating vascular permeability in vitro (12–14). Since it is possible that vascular permeability is determined by the balance between permeability-inducing and -inhibiting factors, we therefore defined the antipermeability activity as the expression level of VEGF subtracted from that of GDNF in a given setting, where the cells treated with a vehicle were used as a control and defined as 100% in PCR analysis. According to this concept, treatments with ATRA decreased VEGF mRNA expression and significantly increased the antipermeability activity (GDNF-VEGF) in U373MG cells (Fig. 1D).
The effects of endogenous retinoic acids are achieved primarily by two retinoic acid isomers (ATRA and 9cRA) and are mediated by two classes of nuclear receptors, RARs (α, β, and γ) and their heterodimeric counterparts retinoid-X receptors (RXRα, -β, and -γ) (16,19). We next examined GDNF expression by using early passages of human astrocytes in the presence of various synthetic retinoids to identify the retinoic acid nuclear receptor subtype(s) responsible for GDNF induction. Treatments with RAR pan-agonists (such as ATRA) and RARα agonists (such as Am580) (20,21) satisfactorily induced GDNF mRNA (Fig. 1E–G), whereas RXR pan-agonists (such as 9cRA and Hx630) did not show marked effects on GDNF when compared with RARα-specific agonists (data not shown). In addition, GFAP expression was significantly induced by the treatment with RARα agonists (Fig. 1E), indicating that RARα stimulants acted on glial cells as a differentiation inducer. Human astrocytes exclusively expressed RARα and -β (Fig. 1H), partially supporting the differential sensitivity of retinoic acids on astrocytes and the idea that RARα stimulants preferentially acted on glial cells to upregulate GDNF expression. This observation was consistent with the published result showing that RARα agonists were, at least in part, important regulators of GDNF expression (17). On the other hand, treatment with RARα stimulants downregulated VEGF expression (Fig. 1E) and were effective in increasing the antipermeability activities, being six times higher in the cells without retinoic acid treatment (Fig. 1I).
RARα transcriptionally stimulates GDNF expression.
ATRA has been shown to trans-activate multiple signal transduction molecules, including p300/CREB (cyclic AMP response element) binding protein (CBP) signal transducer and activator of transcription-3 (STAT3), which eventually play important roles in glial cell differentiation (22–25). We thus examined the alteration in expression of genes belonging to the p300/CBP-STAT3 signal transduction pathway and found that treatments with ATRA and Am580 induced marked increases in the levels of p300/CBP, STAT3, smad1, Notch, Hes-1, and Hes-5 mRNA in glial cells (Fig. 2A). To explore the possible mechanism responsible for the retinoic acid–mediated GDNF upregulation, we isolated an ∼1.8-kb putative promoter fragment including the transcription initiation codon and ∼1.2-kb promoter fragment that lacked all p300 binding motifs and cloned these cDNAs into a luciferase reporter gene plasmid (Fig. 2B, left panel). Luciferase assay revealed that while ATRA and Am580 each exhibited enhanced activity for the full-length promoter, they were ineffective in the deletion mutant lacking the three p300 binding motifs (Fig. 2B, right panel). The percentages of decreases in luciferase activity were 67.0% in ATRA and 93.7% in Am580, compared with the cells transfected with the nondeletion construct (P < 0.05). In addition, ChIP assay demonstrated that p300 was selectively recruited to the GDNF promoter after the treatments with ATRA and Am580 (Fig. 2C). These observations suggested that the expression of GDNF was regulated through the recruitment of an RARα-driven trans-acting coactivator to the ∼1.8-kb 5′-flanking fragment of the GDNF promoter. Although we could not demonstrate a role for the p300/CBP-STAT3 signaling pathway in VEGF downregulation, we cannot rule out its involvement in this process.
RARα-induced phenotypic alteration of glial cells affects endothelial barrier function in vitro.
Our previous study demonstrated that GDNF secreted from glial cells was a critical player in regulating the vascular permeability of the BRB (12–14). To examine whether the gene expression alterations in glial cells induced by RARα agonists modulated the barrier function of endothelial cells, we measured fluxes of radiolabeled molecules using [14C]inulin in a bMVEC-B. This system contained primary microvascular cells derived from bovine brain tissue for the study of microvascular endothelial cell tight junctions and the transport of molecules across the blood-brain barrier, having a histological architecture similar to the BRB (1,12–14). After verifying that the endothelial cells expressed GDNF receptor GFRα1 (Fig. 3A), we treated the endothelium with GDNFr-p and found that it significantly enhanced the barrier function (Fig. 3B). Considering the physiological relevance of our experiment to study the function of the BRB, which is a biological unit comprised of glial cells and capillary endothelium, we cultured glial cells in the lower chamber of a transwell and combined them with an endothelial monolayer that was grown to confluence in the upper chamber in a coculture experiment. Endothelial cells that were cocultured with glial cells pretreated with ATRA and Am580 showed a significant increase of barrier function (Fig. 3C, left panel). Although ATRA was reported to have the ability to enhance the endothelial barrier (26), ATRA alone did not have any effect on the endothelial cells (Fig. 3B), supporting our hypothesis that glial cell–derived cytokines affect the endothelial cells to alter the barrier function in a paracrine manner. Importantly, treatment with GDNF-specific siRNA, which effectively silenced the constitutively expressed GDNF in glial cells, completely inhibited the RARα-mediated enhancement of the endothelial barrier functions (Fig. 3C), strongly suggesting a direct mechanistic link between GDNF expression in glial cells and the regulation of the vascular permeability of the endothelial cells.
Consistent with our observations (Fig. 3B and C), the expression levels of claudin-5, an endothelial tight junction protein and a major determinant of vascular permeability, occludin, and zonula occludens-1 were increased (Fig. 3D), and they were concentrated along the cell-cell borders (Fig. 3E) after the treatment with GDNFr-p. This observation suggested that RARα stimulants regulated barrier function through modulation of expression of a number of tight junction–associated genes. There appeared to be a discrepancy between the results of the RT-PCR assay and immunohistochemistry; however, this might primarily have been due to the lack of commercially available bovine antibodies against tight junction proteins.
RARα agonist is efficient for significant reductions of vascular leakage in vivo.
To further demonstrate the functional significance of glial GDNF, we next evaluated whether RARα stimulants inhibited the vascular leakage in murine diabetic retinopathy. After confirming diabetes in mice (Fig. 4A), we found that diabetic mice had significantly higher expression of both GDNF and VEGF (Fig. 4B and C). While VEGF protein showed significant decreases after the treatments with ATRA and Am580, these treatments did not change the protein levels of GDNF in whole lysates of eyes of diabetic animals (Fig. 4B and C). We cannot currently explain this observation. One possibility is that GDNF expression was induced by compensatory or reactive mechanisms in response to the upregulated VEGF in diabetic animals (5–8). Alternatively, a certain type of advanced glycation end product might have some as of yet unidentified effects on glial cells. However, these alterations in gene expression resulted in a significant increase of antipermeability activities, showing that the RARα stimulation induced apparently opposite trends in endothelial permeability (Fig. 4D). In addition, the altered phenotype mediated by RARα was sufficient for inhibiting vascular leakage to maintain vascular integrity in the retinal microenvironment (Fig. 4E and F). The changes in expression of GDNF and VEGF in diabetic mice were consistent with the results obtained for human vitreous fluid from patients with diabetic retinopathy, showing that the antipermeability activity was significantly decreased in diabetic patients (Fig. 5A and B).
DISCUSSION
Accumulated evidence has suggested that molecular processes involved in vascular growth and vascular hyperpermeability are based on the inappropriate regulation of VEGF (3,4). VEGF thus represents an important target for therapeutic intervention in diabetic retinopathy, and molecular pharmacology that directly inhibits the actions of VEGF has shown considerable promise but has not proven to be satisfactorily effective in blocking the pathology and development of microangiopathies (5–9). Since VEGF is important for maintaining endothelial physiology (27), VEGF suppression therapy may produce unidentified adverse effects on the neural retina. Therefore, a better understanding of the molecular events in vascular alterations is of clinical importance to identify more precise therapeutic targets for diabetic retinopathy.
Indeed, our previous studies have demonstrated that GDNF secreted from glial cells is a critical factor in regulating the vascular permeability of the BRB in a biological unit comprised of capillary endothelial cells and glial cells. In addition, the present study clearly demonstrated that RARα stimulants acted on glial cells, resulting in enhanced expression of GDNF and reciprocal downregulation of VEGF, causally limiting vascular permeability by modulating the tight junction function of the BRB-forming capillary endothelium. Furthermore, phenotypic transformation of glial cells mediated by RARα was sufficient for significant reductions of vascular leakage in diabetic retinopathy. Since the net balance of each participant such as VEGF (a permeability-promoting factor) and GDNF (a permeability-inhibiting factor) is important in diabetic retinopathy, it is acceptable to define the antipermeability activity as the expression level of VEGF subtracted from that of GDNF. Since GDNF is a 2.1 kDa protein and is not detectable in the serum in patients with diabetes (28), it is possible that GDNF is not able to permeate across the BRB. This explanation supports the possibility that the GDNF in the mouse eye originated from glial cells in the retinal microenvironment.
There was no apparent retinoic acid response element on the GDNF or VEGF promoter. However, RARα agonists could significantly alter the gene expression of GDNF and VEGF, and ATRA and Am580 trans-activated many signal transduction molecules, including p300/CBP and STAT3, which are known to play important roles in glial cell differentiation (22,23). In a search to find the putative nuclear factors responsible for GDNF expression, activator protein 1 and stimulatory protein-1 binding motifs were also found in both the GDNF and VEGF promoters, which are reported to be trans-activated by a certain type of retinoic acid (29). Therefore, it is feasible that retinoic acids favor multiple signal transduction components important for glial cell differentiation, associating with direct expression modulation in GDNF and VEGF mRNA at the level of the transcriptional machinery.
RARα stimulation of glial cells effectively inhibited the paracellular permeability of endothelial cells in a paracrine manner. We showed that RARα agonists regulated barrier function by modulating the expression of tight junction proteins such as claudin-5, occludin, and zonula occludens-1, although we cannot explain the mechanism by which GDNF induces the alterations in expression of tight junction–associated genes in endothelial cells. One possible explanation is based on evidence showing that GDNF affects a wide variety of signaling pathways, including those of phosphatidylinositol 3-kinase/Akt, extracellular signal–regulated kinase, p38 mitogen–activated protein kinase, and c-Jun NH2-terminal kinase, which are known to be activated by rearrangement during transfection (RET). Ligation of GDNF to the GDNF-specific receptor GFRα1 leads to dimerization of RET tyrosine kinase (30–32); however, it was somewhat surprising that RET was not expressed in brain microvessels (33). Although further work is needed to address the detailed signal transduction pathway(s) in the endothelial cells in the event of GDNF stimulus, our observations provide important evidence that glial cell–derived cytokines such as GDNF causally limit vascular permeability by modulating the tight junction functions of capillary endothelial cells.
Our results have revealed the functional diversity of the retinal glia and that pharmacological modulation of glial cells serves as a critical barrier-protective mechanism against the development of diabetic retinopathy by regulating endothelial integrity. Given the functional roles of the neural retina in sensing of cellular derangement owing to various metabolic aspects of diabetes, retinoic acids protect against BRB breakdown by suppressing activated phenotypes of glial cells, which potentially provide intraretinal sources of cytokines and chemokines. Since the ocular outcome results from an imbalance of pro- and antipermeable factors, we believe that GDNF is a critical contributing factor for suppressing vascular hyperpermeability in the pathology of retinopathy. While more complex underlying mechanisms appear to be involved in the pathogenesis of diabetic retinopathy, we could successfully clarify that RARα stimulants preferentially inhibited vascular leakiness not only in vitro but also in vivo (Fig. 6). Future studies along this line will pave the way to understanding a wide variety of actions of glial cell–derived cytokines in limiting the progression of diabetic retinopathy. We believe that our results show the potential feasibility of glial cell–targeting therapies for diabetic retinopathy, and these discoveries and subsequent human clinical studies will improve the outcome of diabetes.
RARα agonist stimulates GDNF expression in glial cells. A and B: RT-PCR analysis showing that ATRA upregulates GDNF mRNA expression in a time- and dose-dependent manner in U373MG cells. C: RT-PCR analysis of GDNF in U373MG cells treated with 100 nmol/l ATRA in medium supplemented with endogenous retinoic acid–depleted FBS. D: Net balance of permeability regulating factor (GDNF-VEGF mRNA) in U373MG cells after treatment with 100 nmol/l ATRA. RT-PCR (E), real-time PCR (F), and ELISA (G) analysis showing that expression of GDNF, GFAP, and VEGF is modulated in human astrocytes after treatments with 100 nmol/l ATRA and 10 nmol/l Am580. H: RT-PCR analysis of RARα, -β, and -γ expression in human astrocytes. I: Net balance of permeability regulating factor (GDNF-VEGF mRNA) in human astrocytes after treatment with 100 nmol/l ATRA and 10 nmol/l Am580. The cells treated with DMSO (vehicle) were used as a control (Ctrl), and gene expression in these cells was defined as 100%. *P < 0.05, **P < 0.01 vs. control cells.
RARα agonist stimulates GDNF expression in glial cells. A and B: RT-PCR analysis showing that ATRA upregulates GDNF mRNA expression in a time- and dose-dependent manner in U373MG cells. C: RT-PCR analysis of GDNF in U373MG cells treated with 100 nmol/l ATRA in medium supplemented with endogenous retinoic acid–depleted FBS. D: Net balance of permeability regulating factor (GDNF-VEGF mRNA) in U373MG cells after treatment with 100 nmol/l ATRA. RT-PCR (E), real-time PCR (F), and ELISA (G) analysis showing that expression of GDNF, GFAP, and VEGF is modulated in human astrocytes after treatments with 100 nmol/l ATRA and 10 nmol/l Am580. H: RT-PCR analysis of RARα, -β, and -γ expression in human astrocytes. I: Net balance of permeability regulating factor (GDNF-VEGF mRNA) in human astrocytes after treatment with 100 nmol/l ATRA and 10 nmol/l Am580. The cells treated with DMSO (vehicle) were used as a control (Ctrl), and gene expression in these cells was defined as 100%. *P < 0.05, **P < 0.01 vs. control cells.
p300/CBP-STAT3 pathway is associated with the RARα-induced GDNF expression. A: RT-PCR analysis showing that the treatments with 100 nmol/l ATRA and 10 nmol/l Am580 upregulate genes belonging to the p300/CBP-STAT3 signal transduction pathway. B: Transcriptional analysis of GDNF promoter. Schematic presentation of human proximal GDNF promoter and two constructs having various proximal regions linked to luciferase cDNA (left panel). Promoter activities were examined on HeLa cells in the presence or absence of a vehicle (DMSO), 100 nmol/l ATRA, or 10 nmol/l Am580 (right panel). The cells treated with the vehicle were used as a control (Ctrl) and defined as 100%. Data represent means ± SD from at least three independent experiments, each in triplicate wells. C: ChIP assay of GDNF promoter. Occupancy dynamics of the GDNF promoter with p300 after treatments with 100 nmol/l ATRA and 10 nmol/l Am580 in U373MG cells. Three p300 binding sites (1, 2, and 3) correspond to putative p300 binding motifs in the GDNF promoter (B, left panel). **P < 0.01 vs. control cells.
p300/CBP-STAT3 pathway is associated with the RARα-induced GDNF expression. A: RT-PCR analysis showing that the treatments with 100 nmol/l ATRA and 10 nmol/l Am580 upregulate genes belonging to the p300/CBP-STAT3 signal transduction pathway. B: Transcriptional analysis of GDNF promoter. Schematic presentation of human proximal GDNF promoter and two constructs having various proximal regions linked to luciferase cDNA (left panel). Promoter activities were examined on HeLa cells in the presence or absence of a vehicle (DMSO), 100 nmol/l ATRA, or 10 nmol/l Am580 (right panel). The cells treated with the vehicle were used as a control (Ctrl) and defined as 100%. Data represent means ± SD from at least three independent experiments, each in triplicate wells. C: ChIP assay of GDNF promoter. Occupancy dynamics of the GDNF promoter with p300 after treatments with 100 nmol/l ATRA and 10 nmol/l Am580 in U373MG cells. Three p300 binding sites (1, 2, and 3) correspond to putative p300 binding motifs in the GDNF promoter (B, left panel). **P < 0.01 vs. control cells.
RARα-mediated gene expression alteration in glial cells is sufficient to promote endothelial barrier function in bMVEC-B. A: RT-PCR analysis of GFRα1. B: Treatment with 1 ng/ml GDNFr-p, but not 100 nmol/l ATRA, upregulates barrier function of tight junctions. C: Barrier function of tight junctions cocultured with human astrocytes treated with 100 nmol/l ATRA or 10 nmol/l Am580 in combination with 150 nmol/l GDNF-specific siRNA (left panel). siRNA was transfected 48 h before the assays, and RT-PCR and ELISA analysis were performed to verify the silencing effect of siRNA (right panel). N/D, not detectable. RT-PCR analysis (D) and immunohistochemistry (E) showing the claudin-5 (Cl-5), occludin (Oc), and zonula occludens-1 (ZO-1) expression after treatment with 1 ng/ml GDNFr-p. Signal intensity of control (Ctrl) was defined as 100% and densitometric analysis is also shown below each gel image. Scale bar, 25 μm. *P < 0.05, **P < 0.01 vs. control cells.
RARα-mediated gene expression alteration in glial cells is sufficient to promote endothelial barrier function in bMVEC-B. A: RT-PCR analysis of GFRα1. B: Treatment with 1 ng/ml GDNFr-p, but not 100 nmol/l ATRA, upregulates barrier function of tight junctions. C: Barrier function of tight junctions cocultured with human astrocytes treated with 100 nmol/l ATRA or 10 nmol/l Am580 in combination with 150 nmol/l GDNF-specific siRNA (left panel). siRNA was transfected 48 h before the assays, and RT-PCR and ELISA analysis were performed to verify the silencing effect of siRNA (right panel). N/D, not detectable. RT-PCR analysis (D) and immunohistochemistry (E) showing the claudin-5 (Cl-5), occludin (Oc), and zonula occludens-1 (ZO-1) expression after treatment with 1 ng/ml GDNFr-p. Signal intensity of control (Ctrl) was defined as 100% and densitometric analysis is also shown below each gel image. Scale bar, 25 μm. *P < 0.05, **P < 0.01 vs. control cells.
RARα-mediated phenotypic alteration of glial cells is sufficient to attenuate the BRB breakdown. A: Blood glucose (BS) and urinal sugar (US) in diabetic mice. Urinal sugar was assessed as follows: score 0, negative (−); score 1, slightly positive (±); score 2, weakly positive (+); score 3, moderately positive (++); and score 4, strongly positive (+++). ELISA (B) and Western blot (C) analysis to demonstrate the GDNF and VEGF expression in the mouse eye. Signal intensity of control (Ctrl) was defined as 100%, and densitometric analysis is also shown below each gel image. D: Net balance of permeability regulating factors (GDNF-VEGF protein) in the diabetic mouse eye. FITC leakage from the retina was assessed by laser-scanning confocal microscopy (E) and quantification of FITC (F) in diabetic mice treated with the vehicle (DM), ATRA (DM+ATRA), or Am580 (DM+Am580) compared with the control animals (no diabetes). FITC leakage is clearly observed in diabetic mice, and treatment with ATRA and Am580 protects the retina from diabetic insults. Scale bars, 100 μm. *P < 0.05, **P < 0.01 vs. mice without diabetes, ***P < 0.01 vs. mice without the treatment.
RARα-mediated phenotypic alteration of glial cells is sufficient to attenuate the BRB breakdown. A: Blood glucose (BS) and urinal sugar (US) in diabetic mice. Urinal sugar was assessed as follows: score 0, negative (−); score 1, slightly positive (±); score 2, weakly positive (+); score 3, moderately positive (++); and score 4, strongly positive (+++). ELISA (B) and Western blot (C) analysis to demonstrate the GDNF and VEGF expression in the mouse eye. Signal intensity of control (Ctrl) was defined as 100%, and densitometric analysis is also shown below each gel image. D: Net balance of permeability regulating factors (GDNF-VEGF protein) in the diabetic mouse eye. FITC leakage from the retina was assessed by laser-scanning confocal microscopy (E) and quantification of FITC (F) in diabetic mice treated with the vehicle (DM), ATRA (DM+ATRA), or Am580 (DM+Am580) compared with the control animals (no diabetes). FITC leakage is clearly observed in diabetic mice, and treatment with ATRA and Am580 protects the retina from diabetic insults. Scale bars, 100 μm. *P < 0.05, **P < 0.01 vs. mice without diabetes, ***P < 0.01 vs. mice without the treatment.
GDNF and VEGF expression in the human eye. ELISA analysis showing GDNF and VEGF expression in vitreous fluid (A) and the net balance of permeability regulating factors (GDNF-VEGF protein) (B) in patients with diabetes. *P < 0.05, **P < 0.01 vs. patients without diabetes. Ctrl, control; DM, diabetic.
GDNF and VEGF expression in the human eye. ELISA analysis showing GDNF and VEGF expression in vitreous fluid (A) and the net balance of permeability regulating factors (GDNF-VEGF protein) (B) in patients with diabetes. *P < 0.05, **P < 0.01 vs. patients without diabetes. Ctrl, control; DM, diabetic.
RARα-mediated phenotypic transformation of glial cells antagonizes the loss of tight junction (TJ) integrity induced by diabetes. Given the pleiotropic effects of cellular events and various metabolic abnormalities under hyperglycemia, the retinal glia is a good candidate target cell for these stresses. RARα agonists normalize the physiologically altered phenotypes of glial cells, resulting in the induction of GDNF and a reciprocal decrease of VEGF, which is sufficient to attenuate BRB breakdown and causally maintain vascular integrity in the diabetic retina. Our observations imply that the glial cell is a possible therapeutic target in diabetic retinopathy.
RARα-mediated phenotypic transformation of glial cells antagonizes the loss of tight junction (TJ) integrity induced by diabetes. Given the pleiotropic effects of cellular events and various metabolic abnormalities under hyperglycemia, the retinal glia is a good candidate target cell for these stresses. RARα agonists normalize the physiologically altered phenotypes of glial cells, resulting in the induction of GDNF and a reciprocal decrease of VEGF, which is sufficient to attenuate BRB breakdown and causally maintain vascular integrity in the diabetic retina. Our observations imply that the glial cell is a possible therapeutic target in diabetic retinopathy.
Published ahead of print at http://diabetes.diabetesjournals.org on 25 February 2007. DOI: 10.2337/db06-1431.
Additional information for this article can be found in an online appendix at http://dx.doi.org/10.2337/db06-1431.
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.
Article Information
This study was supported by grants from the Ministry of Education, Culture, Sports, Science and Technology of Japan and the Japan Diabetes Foundation.
We thank Kim Barrymore for help with this manuscript, the Animal Care Facility of Sapporo Medical University, and Dr. Hiroyuki Kagechika (School of Biomedical Sciences, Tokyo Medical and Dental University, Tokyo, Japan) for the gift of various synthetic retinoids.