OBJECTIVE—Suppressors of cytokine signaling (SOCS) are implicated in the etiology of diabetes, obesity, and metabolic syndrome. Here, we show that some SOCS members are induced, while others are constitutively expressed, in retina and examine whether persistent elevation of SOCS levels in retina by chronic inflammation or cellular stress predisposes to developing insulin resistance in retina, a condition implicated in diabetic retinopathy.

RESEARCH DESIGN AND METHODS—SOCS-mediated insulin resistance and neuroprotection in retina were investigated in 1) an experimental uveitis model, 2) SOCS1 transgenic rats, 3) insulin-deficient diabetic rats, 4) retinal cells depleted of SOCS6 or overexpressing SOCS1/SOCS3, and 5) oxidative stress and light-induced retinal degeneration models.

RESULTS—We show that constitutive expression of SOCS6 protein in retinal neurons may improve glucose metabolism, while elevated SOCS1/SOCS3 expression during uveitis induces insulin resistance in neuroretina. SOCS-mediated insulin resistance, as indicated by its inhibition of basally active phosphoinositide 3-kinase/AKT signaling in retina, is validated in retina-specific SOCS1 transgenic rats and retinal cells overexpressing SOCS1/SOCS3. We further show that the SOCS3 level is elevated in retina by oxidative stress, metabolic stress of insulin-deficient diabetes, or light-induced retinal damage and protects ganglion cells from apoptosis, suggesting that upregulation of SOCS3 may be a common physiologic response of neuroretinal cells to cellular stress.

CONCLUSIONS—Our data suggest two-sided roles of SOCS proteins in retina. Whereas SOCS proteins may improve glucose metabolism, mitigate deleterious effects of inflammation, and promote neuroprotection, persistent SOCS3 expression caused by chronic inflammation or cellular stress can induce insulin resistance and inhibit neurotrophic factors, such as ciliary neurotrophic factor, leukemia inhibitory factor, and insulin, that are essential for retinal cell survival.

Insulin resistance and chronic inflammation are implicated in the pathogenesis of diabetic retinopathy, a glucose-mediated microvascular disease that derives, in part, from the inability of retina to adapt to metabolic stress (13). Clinical evidence for a role of insulin resistance as a risk factor for diabetic retinopathy comes from several clinical trials, including the Diabetic Control and Complications Trial and the EURODIAB Prospective Complications Study (4,5). In insulin-deficient diabetic rats, progressive diminution of insulin receptor activity and a marked decrease in insulin receptor substrate (IRS) in retina precede appearance of early features of diabetic retinopathy and neuronal cell death (6). On the other hand, early features of diabetic retinopathy, such as vascular permeability, edema, and increase in inflammatory proteins, are hallmarks of ocular inflammation (1). Although a direct connection between chronic inflammation and insulin resistance in the etiology of diabetic retinopathy has yet to be established, studies on obesity or metabolic syndrome provide a paradigm for understanding the roles of these disparate processes in diabetic retinopathy (7,8). In hepatocytes or adipocytes, activation of insulin receptors recruits IRS proteins to the receptor complex, leading to activation of phosphatidylinositol 3-kinase (PI3K) and AKT signaling pathways, increased glucose metabolism, and cell growth (7). However, several reports (911) have shown that induction of suppressors of cytokine-signaling (SOCS) proteins by inflammatory cytokines desensitizes insulin signaling by targeting IRS1/IRS2 for degradation, leading to development of obesity, hepatic steatosis, or metabolic syndrome. It is therefore of note that IRS2 is essential for insulin signaling in retina (12), and SOCS proteins are elevated in retina during intraocular inflammatory disease (uveitis) (13), suggesting that SOCS-mediated degradation of IRS2 in retina may inhibit signaling downstream of the insulin receptor, providing a mechanistic link between inflammation, insulin resistance, and diabetic retinopathy.

SOCS proteins regulate intensity and duration of cytokine/growth factor signals and integrate multiple extracellular signals that may converge on target cells. The eight-member SOCS family of proteins attenuates cytokine signals through interactions with cytokine/growth factor receptors and signaling proteins, leading to proteosomal degradation of the receptor complex (1416). SOCS proteins are rapidly induced in many cell types in response to cytokines (interferon [IFN]-γ, interleukin [IL]-1β, and IL-6) or growth factors (ciliary neurotrophic factor, leukemia inhibitory factor, fibroblast growth factor, and insulin), and their effects are transient due to their short half-life (1719). However, constitutive SOCS expression occurs in some tissues due to unabated stimulation by chronic inflammation or cellular stress, leading to silencing of critical cellular pathways and predisposition to organ-specific disease development.

In studies on experimental autoimmune uveitis (EAU), a model of human uveitis, we showed that SOCS1 and SOCS3 are induced in retina during uveitis (13,20). Therefore, in this study, we tested the hypothesis that SOCS proteins induced during uveitis can induce insulin resistance and inhibit prosurvival insulin signaling pathways in retina. In addition to intraocular inflammation, we found that factors that induce cellular stress (hypoxia, metabolic stress of diabetes) or photoreceptor damage (high intensity) induce SOCS3 expression in retina, confer partial protection to neuronal cells against apoptosis, and also desensitize insulin signaling in retina cells. We show for the first time that the SOCS6 protein is constitutively expressed in retinal neurons and may function to improve glucose metabolism in neuroretina. Together, these results suggest two-sided roles of SOCS proteins in retina. Benefits of SOCS-mediated protective adaptive responses to inflammation, light damage, hypoxia, or metabolic stress come at a cost, as persistent SOCS1/SOCS3 expression induced by uveitis or chronic stress can induce insulin resistance and trigger diabetic retinopathy.

Animals and induction of diabetes.

C57BL/6 mice were from The Jackson Laboratory (Bar Harbor, ME). For induction of diabetes, overnight-fasted Sprague-Dawley rats (Charles River Laboratories, Wilmington, MA) were given a single intravenous injection of streptozotocin (STZ) (55–65 mg/kg; Sigma) in freshly dissolved 10 mmol sodium citrate (pH 4.5). Diabetes was confirmed by blood glucose >250 mg/dl, and tissues were harvested 6 and 8 weeks after induction. Transgenic rat with targeted expression of SOCS1 using an opsin promoter element was generated and characterized as described (21). Mice/rats were housed in accordance with National Institutes of Health (NIH) guidelines on animal care.

Rat retina, organ culture, culture of human cell lines, and hypoxia studies.

Rat (postnatal day 2 pups) organ cultures were prepared as described (22). Retina organs were stimulated with 100 units/ml IFN-γ (Pepro Tech, Rocky Hill, NJ) and/or 10 nmol/l insulin in Dulbecco's modified Eagle's medium (DMEM) at 37°C, 5% CO2. Human Müller cell line (MI0-M1) (provided by G. Astrid Limb, London, U.K.) was cultured as described (23). Human retinal pigment epithelium cell line (ARPE-19) was cultured as described (24). Before stimulation, cells were placed in serum-free medium for 12 h and then stimulated with 100 units/ml IFN-γ (Pepro Tech), 10 ng/ml IL-4, or 10 nmol/l insulin (Roche). For hypoxia experiments, cells were placed in a hypoxic incubator (37°C, 1% O2, 5% CO2) for various amounts of time (0, 2, 12, or 24 h) as described (25). All experiments were done using cells within the first five passages, and under our experimental condition the cells were under hypoxic conditions after 2 h, as established by induction of hypoxia-inducible factor (HIF)-1α expression (26). Observation of cells by light microscopy after 24 h of hypoxia demonstrated that they were viable and healthy

EAU induction.

We induced EAU in C57BL/6 mice by active immunization with 150 μg bovine interphotoreceptor retinoid binding protein (IRBP) and 300 μg human IRBP peptide (120) in complete Freund's adjuvant containing the Mycobacterium tuberculosis strain H37RA (2.5 mg/ml) (13). Eyes for histology were harvested 0, 14, and 21 days postimmunization, fixed in 10% buffered formalin. Sections were stained with hematoxylin and eosin.

Light treatment.

Mice were dark-adapted overnight and then exposed to white light (5,000 lux) for 6–24 h. Each animal was housed in a separate well-ventilated transparent plastic cage so that one animal could not hide behind another. Temperature was kept at 25.0°C ± 1.0°C during light exposure.

Confocal microscopy.

Sections blocked in 5% normal goat serum were incubated with rabbit polyclonal SOCS6 antibody (Zymed, San Francisco, CA). SOCS6-expressing cells were detected with Alexa-488 conjugated goat anti-rabbit secondary antibody containing DAPI (1 μg/ml) (Invitrogen, Carlsbad, CA) on a Leica-SP2 laser-scanning confocal microscope (Leica Microsystems, Exton, PA) as described (20).

RT-PCR and quantitative RT-PCR analysis.

RNA samples were DNA free. cDNA was generated as described previously (17); each gene-specific primer pair used for RT-PCR analysis spans at least an intron. Quantitative RT-PCR (qRT-PCR) analysis was performed, as previously described (20), using primers and probes from Applied Biosystems. mRNA expression levels were normalized to levels of β-actin and GAPDH housekeeping genes. RT-PCR primers can be found in the online appendix (available at http://dx.doi.org/10.2337/db07-1761).

Western blot analyses.

Preparation of whole-cell lysates from retina, organ cultures, or cell lines and Western blotting were as described (13). Blots were probed with antibodies specific to SOCS1, SOCS3, and SOCS6 (Zymed Laboratories, Inc., South San Francisco, CA); β-actin; p85; SOCS5 and SOCS7; forkhead transcription factor (FOXO) 1; signal transducer and activator of transcription (STAT) 3; vascular endothelial growth factor (VEGF) (Santa Cruz Biotechnology, Santa Cruz, CA); phosphorylated STAT1 (pSTAT1) and pSTAT3; AKT and phosphorylated AKT (pAKT); and IRS-1 and IRS-2 (Cell Signaling, Beverly, MA). Signals were detected using an electrochemiluminescence system (Amersham, Arlington Heights, IL). Some blots were analyzed with the NIH Image Quant program. Each band was normalized to corresponding β-actin band and expressed in arbitrary relative protein expression units.

Ribonuclease protection assay.

A ribonuclease protection assay (RPA) was performed with RNA (10 μg), [α-32P]-UTP radiolabeled RNA probes, and a human SOCS RPA kit (BD Biosciences, San Diego, CA), as recommended by the manufacturer.

Small interfering RNA–mediated silencing of SOCS6 expression.

Cells were cultured for 2–3 days to 60–70% confluency and electroporated with small interfering RNA (siRNA) oligonucleotides (Dharmacon, Lafayette, CO) by Amaxa Nucleofector Technology (Amaxa, Cologne, Germany), according to manufacturer's instructions. Cells were replated in six-well plates and maintained in medium for 48 h before stimulation with agonists.

Cytotoxic assay for detection of apoptosis in mock- and SOCS3-transfected retinal cells.

Apoptosis was induced in ganglion cells (1 × 106 cells/ml) transfected with empty-vector or SOCS3 cDNA with a Nucleofector kit (Amaxa) by culturing for 1 and 3 h in medium containing 1 μmol/l staurosporin (Sigma). Cell death was assessed by fluorescence-activated cell sorter analysis of annexin-V –or 7-amino-actimycin D–labeled cells, according to the manufacturer (BD Biosciences).

Generation of retinal pigment spithelium or retinal ganglion cells overexpressing SOCS1 or SOCS3.

The rat retinal ganglion cell (RGC) line, RGC-5, was generated and characterized as described (27). Construction of SOCS1 or SOCS3 cDNA expression vectors has been described (18,28). RGC-5 and ARPE-19 cells (60–70% confluence) were electroporated by Amaxa Nucleofector Technology.

Statistical analysis.

Experiments were performed at least three times. Figures show data from representative experiments or from combined experiments as indicated. The Student's t test was performed on the data as indicated. P values ≤0.05 are considered statistically significant.

Insulin signaling pathways of the retina are inhibited in EAU.

SOCS1 and SOCS3 are implicated in the etiology of insulin resistance, obesity, and metabolic syndrome (10,29). Here, we have examined whether upregulation of SOCS1/SOCS3 expression in retina during intraocular inflammation (uveitis) predisposes to developing insulin resistance in retina. EAU was induced in C57BL/6 mice by immunization with IRBP, and disease was established by fundoscopy and histology (13,20). Initial clinical signs of EAU appear 7–12 days after immunization, and full-blown clinical disease characterized by IFN-γ secretion develops by postimmunization day 14 (Fig. 1A). Substantial reduction in inflammation is observed by postimmunization day 21 (Fig. 1A), with complete resolution of EAU occurring day 28 postimmunization (13). In line with published reports (13,20), SOCS1 and SOCS3 mRNA expression is induced in retina, with the highest levels detected at peak of the disease (data not shown). Western blot analysis shows that normal mouse retina has high basally active insulin receptor signaling, as indicated by constitutive expression of pAKT, IRS proteins, and p85 regulatory subunit of PI3K in retina (Fig. 1B). These analyses also show that an increase in SOCS3 protein during EAU correlates with decreased PI3K/AKT signaling in EAU retina, as indicated by diminution in levels of pAKT, p85, and IRS proteins (Fig. 1B and C). Increase in SOCS3 expression and decrease in PI3K/AKT signaling are also observed in EAU mouse spleen (Fig. 1D). Of note, the slight decrease in IRS2 in EAU retina is in concert with reports showing that IRS2 is required for survival/insulin signaling in photoreceptor cells (12) and sensitive to SOCS3-mediated degradation (11). These results establish a correlation between upregulation of SOCS1 and SOCS3 in retina during uveitis and diminished biological responses to insulin in retina.

SOCS genes are differentially regulated in the retina by proinflammatory cytokines.

SOCS1 and SOCS3 expression in retina during uveitis is a negative-feedback response that mitigates retinal pathology by limiting intensity and duration of activities of proinflammatory cytokines produced by uveitogenic T-cells (13). However, inhibition of basal insulin/AKT signaling pathways in retina during EAU suggests that SOCS1/SOCS3 may also be induced in retinal cells by inflammatory cytokines. To establish that retinal cells express SOCS genes in response to proinflammatory cytokines, we stimulated ARPE-19 and MI0-M1 retinal cells with IFN-γ or IL-4. qRT-PCR (Fig. 2A) and RPA (Fig. 2B) unambiguously show that exposure of resident retinal cells to proinflammatory cytokines upregulates SOCS1/SOCS3 expression. Of particular interest is constitutive expression of SOCS5, SOCS6, and SOCS7 in human retinal cells (Fig. 2B) as well as mouse and rat retina (Fig. 2C and D). These results suggest that SOCS genes are differentially regulated in retina and may have distinct functions in the physiology of neuroretina. Because SOCS6 has been shown to improve glucose metabolism (30), we examined the spatial localization of SOCS6 protein in retina. We show that SOCS6 is localized to ganglion cell and photoreceptor layers of mouse retina (Fig. 2E) and retinal pigment epithelium (RPE). The anatomic localization of SOCS6 protein to regions of high metabolic demands in the retina is consistent with its role in supporting glucose metabolic pathways.

SOCS1 and SOCS3 induce insulin resistance in retinal cells.

To further examine whether cytokine-induced overexpression of SOCS proteins in retina causes insulin resistance, we exposed postnatal day 2 rat retinal organ cultures to IFN-γ and/or insulin for 12 h. As shown (Fig. 3A), a relatively high level of SOCS1 is induced by IFN-γ, while only a modest level of SOCS3 expression is induced by insulin or IFN-γ (Fig. 3A). However, marked elevation of SOCS3 by IFN-γ occurs in retinal cells concurrently stimulated by insulin (Fig. 3A). We further show that enhanced expression of SOCS1/SOCS3 proteins in retina derives, in part, from synergistic activation of STAT1 by insulin and IFN-γ (Fig. 3C). In concert with high metabolic requirements of retina (6,31), insulin signaling is constitutively active in retina, as indicated by detection of pAKT in freshly isolated primary retinal cells (Fig. 3B). However, pAKT activity of freshly isolated retinal cells is inhibited by IFN-γ, even in our insulin/IFN-γ coculture experiment, where cells are continuously stimulated by exogenous insulin (Fig. 3B, lanes 3 and 4). Potential mechanism(s) by which IFN-γ reduces AKT phosphorylation in both basal and insulin-stimulated states may be similar to those described in liver and adipose cells, where induction of SOCS1/SOCS3 by IL-6 or tumor necrosis factor-α desensitizes insulin signaling by targeting IRS1/IRS2 for degradation (911). Thus, diminution of constitutive pAKT in retinal cells (Fig. 3A) following IFN-γ–induced upregulation of SOCS1 and SOCS3 expression (Fig. 3A) suggests that IFN-γ secretion during uveitis may diminish biological responses of retinal cells to insulin in retina. We have also generated rat ganglion (Fig. 3D) or ARPE-19 (Fig. 3E) cells overexpressing SOCS1 or SOCS3, and either SOCS member suppresses AKT/PI3K activation by insulin, underscoring potential roles of these SOCS proteins in inducing insulin resistance in retinal cells. To directly examine effects of constitutive expression of SOCS1 in retina, we generated transgenic rats with targeted overexpression of SOCS1 in retina (21). Similar to our in vitro data, elevation of SOCS1 level in retina induces insulin resistance in transgenic rat retina as indicated by low basal pAKT activity in freshly isolated retinal cells and diminished ability of insulin to activate AKT pathways in transgenic retina (Fig. 3F).

SOCS3 is upregulated in retina by light damage, oxidative stress, and insulin-deficient diabetes.

Desensitization of retinal cells to insulin signals by SOCS proteins led us to examine other cellular pathways that may cause elevation of SOCS proteins in retina. We therefore examined whether SOCS proteins are induced in retina in response to three distinct types of stress implicated in the development of retinal degenerative diseases: high-intensity light, oxidative stress, and metabolic stress induced by insulin-deficient diabetes. For light-induced retina damage, mice were exposed to high-intensity light for varying amounts of time, ranging from 24 h (Fig. 4A) to 6.5 h (Fig. 4B). RT-PCR (Fig. 4A) and Western blot (Fig. 4B) analysis of mouse retina revealed that exposure of retinal cells to high-intensity light that induces apoptotic death of retina cells in vivo (32) elicits a rapid activation of STAT3, upregulation of VEGF, and SOCS3 expression. In three separate experiments, activation of STAT3/VEGF expression precedes peak expression of SOCS3 (Fig. 4B). Thus, consistent with published reports (33,34), light-induced VEGF may activate STAT3 and induce SOCS3 expression. It may well be that light-induced changes in SOCS3/STAT3 plays a role in protecting photoreceptor cells from light-induced apoptosis. Similarly, culturing retinal cells under hypoxic condition induces VEGF and HIF-1α expression, with HIF-1α expression observed as early as 2 h, while high-level VEGF expression is detected at 12 h (Fig. 4C). On the other hand, genetic studies (35,36) show that STAT3 is required for VEGF and HIF-1α expression, suggesting that VEGF may be regulated by STAT3 and under negative-feedback regulation by SOCS3 in retina. We also investigated whether insulin deficiency in retina is associated with elevation of SOCS proteins in retina. Diabetes was induced by injection of STZ, and by week 8 postinjection rats developed large focal retinal degeneration, retinal folds, and thinning of retinal layers (Fig. 4D). Analyses of STZ-induced diabetic (glucose level >400 mg/dl) or nondiabetic rat retinae reveal that SOCS3 mRNA (Fig. 4E) and protein (Fig. 4F) are substantially elevated in all STZ-induced diabetic rat retina examined. Interestingly, increase in SOCS3 in diabetic retina is accompanied by decrease in pAKT. Remarkably, retinal cells selectively induce SOCS3 but not other SOCS family members (Fig. 4B) in response to light damage, oxidative stress, or diabetes-induced retinal damage, suggesting that SOCS3 may play a role in protecting the neuroretina from cellular stress. In fact, we provide direct evidence that SOCS3 confers protection to retinal cells from apoptosis. Survival of retinal ganglion cells transfected with empty-vector or SOCS3 cDNA was assessed following treatment with the apoptosis-inducing chemical staurosporin. Apoptotic and necrotic cells were assessed by annexin-V and 7-amino-actimycin D staining assays, respectively, and enhanced survival of SOCS3 transfected cells (45.8 vs. 30.8%) and reduced number of SOCS3 transfectants undergoing apoptosis (39.7 vs. 59.9%) after 3 h treatment indicate that SOCS3 protects retinal cells from apoptosis (Fig. 4G).

SOCS6 activates the prosurvival AKT pathway and inhibits the proapoptotic FOXO1 protein.

In the retina, we observe a dichotomous pattern of SOCS expression: whereas SOCS1, SOCS2, SOCS3, and cytokine-induced SH2 protein (CIS) are barely detectable, SOCS5, SOCS6, and SOCS7 are constitutively expressed (Fig. 2C and D). The spatial localization of SOCS6 in ganglion and photoreceptor cell layers is of particular interest as these regions of retina are indispensable components of the visual apparatus and major sites of retinal degenerative changes in diabetic retinopathy. Small interfering RNA (siRNA)-mediated silencing of SOCS6 expression was therefore used to investigate functional relevance of SOCS6 in ocular cells. Optimal condition for efficient siRNA transfection into ARPE-19 cells was established with green fluorescent protein (GFP)-siRNA oligonucleotides (Fig. 5A), and 1.5 μg SOCS6-siRNA was found to induce an 85% reduction of endogenous SOCS6 RNA expression without appreciable effect on cell viability (Fig. 5B). We show that maximum depletion of the endogenous SOCS6 protein that occurs consistently in day 6–transfected cells coincides temporarily with inhibition of insulin-induced activation of AKT (Fig. 5C). Dependence of pAKT signaling on SOCS6 is of functional relevance because this pathway, downstream of activated insulin/IRS2, is essential for retinal cell survival (12). A well-characterized prosurvival pathway induced by insulin/IRS2 derives from obligatory inactivation of FOXO1 by AKT kinases, resulting in inhibition of proapoptotic and growth inhibitory functions of this forkhead family transcription factor (37). We therefore investigated whether SOCS6 is involved in regulating this pathway. Silencing of SOCS6 results in blockage of insulin/PI3K/AKT-mediated inactivation of FOXO1 (Fig. 5D), suggesting a potential role of SOCS6 in promoting insulin-induced survival of retinal cells.

In a previous study (38,39), we showed that transgenic rats with targeted expression of IFN-γ in the retina induced constitutively high levels of SOCS1 and SOCS3 in the retina and by 3 months of age the rats developed severe retinal degeneration. In this study, we have tested the hypothesis that chronic intraocular inflammation or other sources of cellular stress that induce persistent SOCS expression in retina might be risk factors for the development of retinal diseases. Particular emphasis is on the potential role of persistent stimulation of SOCS expression in retina by inflammatory cytokines, oxidative stress, light damage, or metabolic stress in the development of insulin resistance, an important complication of diabetes and contributory factor of the potentially blinding disease diabetic retinopathy.

We show here that induction of SOCS1 and SOCS3 expression during uveitis inhibits signaling downstream of insulin receptor in the retina (Fig. 1B and C), and the reduction in constitutive insulin signaling derives in part from a decrease in levels of pAKT, p85 (regulatory subunit of PI3K), and IRS proteins in the retina. We further show that insulin synergizes with IFN-γ to enhance SOCS1/SOCS3 expression in retinal cells, suggesting that copious amounts of IFN-γ secreted in retina by inflammatory cells during uveitis (13) may synergize with insulin to elevate levels of SOCS1/SOCS3 proteins, resulting in desensitization of retinal cells to insulin signals. The role of SOCS in desensitizing retinal cells to insulin signals is further established by studies showing that forced overexpression of SOCS1 or SOCS3 protein in rat retinal ganglion cells, human RPE cells, or primary rat retinal cells inhibits insulin-induced PI3K/AKT signaling, an important indicator of the development of insulin resistance (Fig. 3). Insulin resistance in fresh retina organ cultures (Fig. 3B and C), in SOCS1- or SOCS3-transfected retinal cells (Fig. 3D and E), or during EAU (Fig. 1) is validated in transgenic rats with targeted overexpression of SOCS1 in the retina (Fig. 3F). It is remarkable that all SOCS1 transgenic rats exhibit low basal insulin/AKT signaling activity and are unable to induce appreciable PI3K/AKT signaling even after stimulation by exogenous insulin. Although SOCS1 and SOCS3 have been implicated in pathogenesis of type 2 diabetes (40), obesity (40), hepatic steatosis (8), and metabolic syndrome (41), this is the first report showing that elevated levels of SOCS proteins during uveitis diminish basally active insulin and AKT signaling in retina.

In addition to inflammation, high-intensity light, oxidative stress, and metabolic stress of insulin-deficient diabetes also induce SOCS3 protein in the retina but not SOCS1, SOCS6, or other SOCS members. As SOCS3 inhibits signals downstream of the insulin receptor in retinal cells (Fig. 3D and E), these results suggest that other physiologic states that elevate SOCS3 levels may contribute to development of insulin resistance in retina. Furthermore, in our rodent models of light-induced retinal degeneration or hypoxia, VEGF, a survival factor for retinal neurons (42,43), is rapidly upregulated in retina, in part through STAT3-dependent mechanisms (Fig. 4B and C), consistent with requirement of STAT3 for transcription of VEGF and HIF-1α mRNA (35,36,44). While VEGF may provide prosurvival functions in retina, accumulation of VEGF in the eye is implicated in the development of retinopathies, and anti-VEGF antibody is effective in treatment of diabetic retinal neovascularization (45). These diametrically polar effects of VEGF in the retina suggest that STAT3-induced VEGF expression is exquisitely regulated in the retina, as activated STAT3 is preferentially localized to neovascular retinal vessels in a mouse model of ischemia-induced retinal neovascularization (46). However, as STAT3 promotes proliferation and prevents apoptosis of some cell types (47), we cannot rule out the possibility that pathways activated in the retina by STAT3 may play a role in protecting photoreceptor cells from light-induced apoptosis. SOCS3 is a major inhibitor of STAT3, and we observed a temporal correlation between STAT3-induced VEGF expression and SOCS3 induction in light-treated retina (Fig. 4B) or ocular cells subjected to oxidative stress (Fig. 4C), suggesting that stress-induced activation of STAT3 and VEGF expression are under negative feedback regulation by SOCS3. Physiological relevance of SOCS3 as a stress response factor is further underscored by the high constitutive SOCS3 mRNA (Fig. 4E) and protein (Fig. 4F) expression in degenerating retina (Fig. 4D) of insulin-deficient STZ-induced diabetic rats and by a recent report showing that loss of SOCS3-mediated feedback regulation converts STAT3 function from antiapoptotic to proapoptotic (47).

In this study, we also investigated possible roles of other SOCS members in insulin signaling. We found that SOCS5, SOCS6, and SOCS7 proteins are constitutively expressed in the retina, suggesting that they may have housekeeping functions in the retina (Fig. 2C–E). Surprisingly, depletion of endogenous SOCS6 in ARPE-16 cells inhibits insulin signaling, suggesting that constitutive SOCS6 expression may serve to maintain high basal insulin/AKT signaling in retina and improve glucose metabolism to meet the high metabolic demands of the retina. This is in line with recent findings (35) indicating that SOCS6 improves glucose metabolism by overcoming the inhibitory effects of p85 monomers on PI3K-dependent signaling pathways. The anatomic localization of SOCS6 expression in the inner retina with sparse vascularity, low oxygen tension, and exclusive dependence on glycolysis or in photoreceptors that are engaged in the high energy–requiring process of phototransduction is consistent with its role in glucose metabolism. Besides potentiation of AKT signaling, SOCS6 may also promote survival of retinal cells. In insulin-sensitive cells, progrowth/prosurvival functions of insulin necessitate inhibition of proapoptotic/growth inhibitory functions of forkhead transcription factors. We show here that insulin-induced inactivation of FOXO1 is inhibited in SOCS6-silenced ARPE-19 cells (Fig. 5D), suggesting that SOCS6 may be involved in regulating FOXO1 in retina.

Retinal dystrophies, including diabetic retinopathy, cause severe vision impairment or blindness, and development of effective treatment will benefit from better understanding of cellular pathways that inhibit neuronal cell death or repair of damaged neurons. Molecular cues that influence the decision to allow death of irreversibly damaged photoreceptors or provide survival signals that rescue partially damaged cells are unknown, and signals from neurotrophic factors that mediate reparative processes in retina are suspected to play important roles. Retinal neurotrophic factors include ciliary neurotrophic factor, leukemia inhibitory factor, and VEGF, and they all mediate their effects by activating Janus kinase/STAT pathways and are under negative-feedback regulation by SOCS proteins. It is therefore remarkable that SOCS3 is induced in retina in response to infection, light damage, oxidative stress, or insulin-deficient diabetes. Data presented thus suggest two-sided roles of SOCS proteins in retina. Whereas transient induction of SOCS3 may be a protective adaptive response to inflammation, light damage, hypoxia, or metabolic stress, persistently high SOCS3 levels in retina is potentially pathogenic because it would inhibit survival signals that emanate from neurotrophic factors. Nonetheless, of the SOCS proteins, SOCS3 might prove to be useful in inhibiting inflammation, VEGF, and angiogenesis, while targeted delivery of SOCS6 to retinal cells may improve glucose metabolism in diabetic subjects and confer protection against metabolic stress.

FIG. 1.

Induction of SOCS1 and SOCS3 during EAU correlates with inhibition of insulin signaling in the retina. A: Six-week-old mouse was immunized with IRBP in complete Freund's adjuvant and 14 days later eyes were enucleated. Four-micron-thick sections were cut through the retina and stained with hematoxylin and eosin. B: Western blot analysis of whole-cell protein extract prepared from the retina (B and C) or spleen (D) of normal mouse or mouse with EAU. B, C, and D (bottom panels): Intensities of the Western blot bands were analyzed in a densitometer to quantify the relative protein expression levels.

FIG. 1.

Induction of SOCS1 and SOCS3 during EAU correlates with inhibition of insulin signaling in the retina. A: Six-week-old mouse was immunized with IRBP in complete Freund's adjuvant and 14 days later eyes were enucleated. Four-micron-thick sections were cut through the retina and stained with hematoxylin and eosin. B: Western blot analysis of whole-cell protein extract prepared from the retina (B and C) or spleen (D) of normal mouse or mouse with EAU. B, C, and D (bottom panels): Intensities of the Western blot bands were analyzed in a densitometer to quantify the relative protein expression levels.

Close modal
FIG. 2.

Expression of SOCS family genes is differentially regulated in the retina. A: Induction of SOCS expression in human retinal cell lines by proinflammatory cytokines was detected by qRT-PCR (A) or RPA (B). C: RT-PCR analysis of SOCS expression in mouse retina: SOCS1 (lane 1), SOCS2 (lane 2), SOCS3 (lane 3), SOCS5 (lane 4), SOCS6 (lane 5), and SOCS7 (lane 6). D: Detection of SOCS5, SOCS6, and SOCS7 proteins in the retina and retinal cell lines by Western blot analysis: rat retina (lane 1), human ARPE-19 cell line (lane 2), and human Müller cell line (lane 3). E: Localization of SOCS6 protein expression in the mouse retina by immunohistochemical analysis: GLC, ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer; Pho, photoreceptors.

FIG. 2.

Expression of SOCS family genes is differentially regulated in the retina. A: Induction of SOCS expression in human retinal cell lines by proinflammatory cytokines was detected by qRT-PCR (A) or RPA (B). C: RT-PCR analysis of SOCS expression in mouse retina: SOCS1 (lane 1), SOCS2 (lane 2), SOCS3 (lane 3), SOCS5 (lane 4), SOCS6 (lane 5), and SOCS7 (lane 6). D: Detection of SOCS5, SOCS6, and SOCS7 proteins in the retina and retinal cell lines by Western blot analysis: rat retina (lane 1), human ARPE-19 cell line (lane 2), and human Müller cell line (lane 3). E: Localization of SOCS6 protein expression in the mouse retina by immunohistochemical analysis: GLC, ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer; Pho, photoreceptors.

Close modal
FIG. 3.

SOCS1 or SOCS3 induces insulin resistance in retinal cells. A: RT-PCR analysis showing synergistic induction of SOCS3 expression in rat retina organ culture by IFN-γ and insulin. Western blot analysis of extracts from rat organ cultures stimulated with insulin and/or IFN-γ (B) or insulin, IL-4, and/or IFN-γ (C). Western blot analysis showing inhibition of insulin-induced pAKT signaling in rat ganglion (D) or human RPE cells overexpressing SOCS1 or SOCS3 (E). F: Western blot analysis showing inhibition of insulin-induced activation of AKT signaling in a transgenic rat strain with targeted overexpression of SOCS1 in the retina. Retina organs were cultured for 12 h in normal serum in the absence (lanes 2 and 4) or presence of 10 nmol/l insulin (lanes 3 and 5). Lane 1, freshly isolated retina from wild-type rat.

FIG. 3.

SOCS1 or SOCS3 induces insulin resistance in retinal cells. A: RT-PCR analysis showing synergistic induction of SOCS3 expression in rat retina organ culture by IFN-γ and insulin. Western blot analysis of extracts from rat organ cultures stimulated with insulin and/or IFN-γ (B) or insulin, IL-4, and/or IFN-γ (C). Western blot analysis showing inhibition of insulin-induced pAKT signaling in rat ganglion (D) or human RPE cells overexpressing SOCS1 or SOCS3 (E). F: Western blot analysis showing inhibition of insulin-induced activation of AKT signaling in a transgenic rat strain with targeted overexpression of SOCS1 in the retina. Retina organs were cultured for 12 h in normal serum in the absence (lanes 2 and 4) or presence of 10 nmol/l insulin (lanes 3 and 5). Lane 1, freshly isolated retina from wild-type rat.

Close modal
FIG. 4.

Retinal cells induce expression of SOCS3 in response to light damage, hypoxia, or insulin-deficient diabetes. A: RT-PCR analysis of mouse retina following 24 h exposure to high-intensity light (5,000 lux). B: Western blot analysis of retina following 6.5 h exposure of mice to high-intensity light (5,000 lux). Right panel: Intensities of the Western blot bands were analyzed in a densitometer to quantify the relative protein expression levels. These results are from three independent experiments. C: Mouse retina explants were propagated for varying amounts of time under hypoxia condition. Induction of VEGF and HIF-1α expression was detected by RT-PCR, while SOCS3 expression was by qRT-PCR analysis. These results are from three independent experiments. D: Histology of rat retina showing development of retinal degeneration in 8-week STZ-induced diabetic rats but not in non–STZ-induced control rat. RT-PCR (note: each lane represents an individual rat) (E) and Western blot (F) analysis of SOCS3 expression in retina of normal or insulin-deficient diabetic rats. Right panel: Intensities of the Western blot bands were analyzed in a densitometer to quantify the relative protein expression levels. G: Annexin V–fluorescein isothiocyanate histograms showing percentage of apoptotic cells following treatment with staurosporin. Necrotic cells were detected by fluorescence-activated cell sorter analysis of 7-amino-actimycin D labeled.

FIG. 4.

Retinal cells induce expression of SOCS3 in response to light damage, hypoxia, or insulin-deficient diabetes. A: RT-PCR analysis of mouse retina following 24 h exposure to high-intensity light (5,000 lux). B: Western blot analysis of retina following 6.5 h exposure of mice to high-intensity light (5,000 lux). Right panel: Intensities of the Western blot bands were analyzed in a densitometer to quantify the relative protein expression levels. These results are from three independent experiments. C: Mouse retina explants were propagated for varying amounts of time under hypoxia condition. Induction of VEGF and HIF-1α expression was detected by RT-PCR, while SOCS3 expression was by qRT-PCR analysis. These results are from three independent experiments. D: Histology of rat retina showing development of retinal degeneration in 8-week STZ-induced diabetic rats but not in non–STZ-induced control rat. RT-PCR (note: each lane represents an individual rat) (E) and Western blot (F) analysis of SOCS3 expression in retina of normal or insulin-deficient diabetic rats. Right panel: Intensities of the Western blot bands were analyzed in a densitometer to quantify the relative protein expression levels. G: Annexin V–fluorescein isothiocyanate histograms showing percentage of apoptotic cells following treatment with staurosporin. Necrotic cells were detected by fluorescence-activated cell sorter analysis of 7-amino-actimycin D labeled.

Close modal
FIG. 5.

siRNA-mediated silencing of SOCS6 expression inhibits insulin-induced activation of AKT signaling in human retinal cells. A: Verification of siRNA transfection efficiency in human RPE cells (ARPE-19) transfected with GFP plasmid and GFP-specific siRNA. B: qRT-PCR analysis of SOCS6 mRNA expression in ARPE-19 days after transfection with GFP-specific (GFP siRNA) or SOCS6-specific (SOCS6 siRNA) siRNA. Western blot analyses showing: inhibition of insulin-induced pAKT signaling (C) or inhibition of FOXO1 inactivation in day 6 SOCS6-siRNA–transfected ARPE-19 (D).

FIG. 5.

siRNA-mediated silencing of SOCS6 expression inhibits insulin-induced activation of AKT signaling in human retinal cells. A: Verification of siRNA transfection efficiency in human RPE cells (ARPE-19) transfected with GFP plasmid and GFP-specific siRNA. B: qRT-PCR analysis of SOCS6 mRNA expression in ARPE-19 days after transfection with GFP-specific (GFP siRNA) or SOCS6-specific (SOCS6 siRNA) siRNA. Western blot analyses showing: inhibition of insulin-induced pAKT signaling (C) or inhibition of FOXO1 inactivation in day 6 SOCS6-siRNA–transfected ARPE-19 (D).

Close modal

Published ahead of print at http://diabetes.diabetesjournals.org on 20 March 2008. DOI: 10.2337/db07-1761.

Additional information for this article can be found in an online appendix at http://dx.doi.org/10.2337/db07-1761.

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.

This research was funded by the Intramural Research Programs of the National Eye Institute (NEI) and the National Institutes of Health (NIH).

The authors thank Drs. Robert Fariss and Jen Yue Tsai (NEI Imaging Core Unit) for assistance with confocal microscopy; Dr. Emmanuel Opara and Sanja Benac (Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, Chicago, Illinois) for assistance in inducing diabetes in rats; Dr. Chandrasekharam Nagineni (NEI, NIH) for providing the RGC-5 ganglion cell line generated by Dr. Neeraj Agarwal (UNT Health Science Center, Fort Worth, Texas); Drs. Ronald A. Bush and Igal Gery (NEI, NIH) for assistance with light damage experiments; and Dr. Jorge Sztein (NEI Genetic Engineering Core unit) and Rashid Mahdi (Laboratory of Immunology, NEI, NIH) for technical assistance in generating and characterizing SOCS1 transgenic rats.

1.
Antonetti DA, Barber AJ, Bronson SK, Freeman WM, Gardner TW, Jefferson LS, Kester M, Kimball SR, Krady JK, LaNoue KF, Norbury CC, Quinn PG, Sandirasegarane L, Simpson IA: Diabetic retinopathy: seeing beyond glucose-induced microvascular disease.
Diabetes
55
:
2401
–2411,
2006
2.
Meleth AD, Agron E, Chan CC, Reed GF, Arora K, Byrnes G, Csaky KG, Ferris FL 3rd, Chew EY: Serum inflammatory markers in diabetic retinopathy.
Invest Ophthalmol Vis Sci
46
:
4295
–4301,
2005
3.
Abu el Asrar AM, Maimone D, Morse PH, Gregory S, Reder AT: Cytokines in the vitreous of patients with proliferative diabetic retinopathy.
Am J Ophthalmol
114
:
731
–736,
1992
4.
Zhang L, Krzentowski G, Albert A, Lefebvre PJ: Risk of developing retinopathy in Diabetes Control and Complications Trial type 1 diabetic patients with good or poor metabolic control.
Diabetes Care
24
:
1275
–1279,
2001
5.
Chaturvedi N, Sjoelie AK, Porta M, Aldington SJ, Fuller JH, Songini M, Kohner EM: Markers of insulin resistance are strong risk factors for retinopathy incidence in type 1 diabetes.
Diabetes Care
24
:
284
–289,
2001
6.
Reiter CE, Wu X, Sandirasegarane L, Nakamura M, Gilbert KA, Singh RS, Fort PE, Antonetti DA, Gardner TW: Diabetes reduces basal retinal insulin receptor signaling: reversal with systemic and local insulin.
Diabetes
55
:
1148
–1156,
2006
7.
Taniguchi CM, Emanuelli B, Kahn CR: Critical nodes in signaling pathways: insights into insulin action.
Nat Rev Mol Cell Biol
7
:
85
–96,
2006
8.
Ueki K, Kadowaki T, Kahn CR: Role of suppressors of cytokine signaling SOCS-1 and SOCS-3 in hepatic steatosis and the metabolic syndrome.
Hepatol Res
33
:
185
–192,
2005
9.
Ueki K, Kondo T, Kahn CR: Suppressor of cytokine signaling 1 (SOCS-1) and SOCS-3 cause insulin resistance through inhibition of tyrosine phosphorylation of insulin receptor substrate proteins by discrete mechanisms.
Mol Cell Biol
24
:
5434
–5446,
2004
10.
Emanuelli B, Peraldi P, Filloux C, Chavey C, Freidinger K, Hilton DJ, Hotamisligil GS, Van Obberghen E: SOCS-3 inhibits insulin signaling and is up-regulated in response to tumor necrosis factor-alpha in the adipose tissue of obese mice.
J Biol Chem
276
:
47944
–47949,
2001
11.
Rui L, Yuan M, Frantz D, Shoelson S, White MF: SOCS-1 and SOCS-3 block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2.
J Biol Chem
277
:
42394
–42398,
2002
12.
Yi X, Schubert M, Peachey NS, Suzuma K, Burks DJ, Kushner JA, Suzuma I, Cahill C, Flint CL, Dow MA, Leshan RL, King GL, White MF: Insulin receptor substrate 2 is essential for maturation and survival of photoreceptor cells.
J Neurosci
25
:
1240
–1248,
2005
13.
Takase H, Yu CR, Liu X, Fujimoto C, Gery I, Egwuagu CE: Induction of suppressors of cytokine signaling (SOCS) in the retina during experimental autoimmune uveitis (EAU): potential neuroprotective role of SOCS proteins.
J Neuroimmunol
168
:
118
–127,
2005
14.
Naka T, Matsumoto T, Narazaki M, Fujimoto M, Morita Y, Ohsawa Y, Saito H, Nagasawa T, Uchiyama Y, Kishimoto T: Accelerated apoptosis of lymphocytes by augmented induction of Bax in SSI-1 (STAT-induced STAT inhibitor-1) deficient mice.
Proc Natl Acad Sci U S A
95
:
15577
–15582,
1998
15.
Sasaki A, Yasukawa H, Shouda T, Kitamura T, Dikic I, Yoshimura A: CIS3/SOCS-3 suppresses erythropoietin (EPO) signaling by binding the EPO receptor and JAK2.
J Biol Chem
275
:
29338
–29347,
2000
16.
Yasukawa H, Misawa H, Sakamoto H, Masuhara M, Sasaki A, Wakioka T, Ohtsuka S, Imaizumi T, Matsuda T, Ihle JN, Yoshimura A: The JAK-binding protein JAB inhibits Janus tyrosine kinase activity through binding in the activation loop.
EMBO J
18
:
1309
–1320,
1999
17.
Egwuagu CE, Yu CR, Zhang M, Mahdi RM, Kim SJ, Gery I: Suppressors of cytokine signaling proteins are differentially expressed in Th1 and Th2 cells: implications for Th cell lineage commitment and maintenance.
J Immunol
168
:
3181
–3187,
2002
18.
Yu CR, Mahdi RM, Ebong S, Vistica BP, Chen J, Guo Y, Gery I, Egwuagu CE: Cell proliferation and STAT6 pathways are negatively regulated in T cells by STAT1 and suppressors of cytokine signaling.
J Immunol
173
:
737
–746,
2004
19.
Samardzija M, Wenzel A, Aufenberg S, Thiersch M, Reme C, Grimm C: Differential role of Jak-STAT signaling in retinal degenerations.
FASEB J
20
:
2411
–2413,
2006
20.
Amadi-Obi A, Yu CR, Liu X, Mahdi RM, Clarke GL, Nussenblatt RB, Gery I, Lee YS, Egwuagu CE: T(H)17 cells contribute to uveitis and scleritis and are expanded by IL-2 and inhibited by IL-27/STAT1.
Nat Med
13
:
711
–718,
2007
21.
Ham DI, Kim SJ, Chen J, Vistica BP, Fariss RN, Lee RS, Wawrousek EF, Takase H, Yu CR, Egwuagu CE, Chan CC, Gery I: Central immunotolerance in transgenic mice expressing a foreign antigen under control of the rhodopsin promoter.
Invest Ophthalmol Vis Sci
45
:
857
–862,
2004
22.
Hofmann H, Schulz-key S, Hertle D, Kirsch M: Orgaotypic cultures of the rat retina. Poindron P, Piguet P, Forster E, Eds. In
New Methods for Culturing Cells From Nervous Tissues
. Basel, Karger,
2005
, p. 
1
:
58
–73 [Bio Valley monograph]
23.
Limb GA, Salt TE, Munro PM, Moss SE, Khaw PT: In vitro characterization of a spontaneously immortalized human Muller cell line (MIO-M1).
Invest Ophthalmol Vis Sci
43
:
864
–869,
2002
24.
Li W, Nagineni CN, Hooks JJ, Chepelinsky AB, Egwuagu CE: Interferon-gamma signaling in human retinal pigment epithelial cells mediated by STAT1, ICSBP, and IRF-1 transcription factors.
Invest Ophthalmol Vis Sci
40
:
976
–982,
1999
25.
Udono T, Takahashi K, Nakayama M, Yoshinoya A, Totsune K, Murakami O, Durlu YK, Tamai M, Shibahara S: Induction of adrenomedullin by hypoxia in cultured retinal pigment epithelial cells.
Invest Ophthalmol Vis Sci
42
:
1080
–1086,
2001
26.
Forooghian F, Razavi R, Timms L: Hypoxia-inducible factor expression in human RPE cells.
Br J Ophthalmol
91
:
1406
–1410,
2007
27.
Krishnamoorthy RR, Agarwal P, Prasanna G, Vopat K, Lambert W, Sheedlo HJ, Pang IH, Shade D, Wordinger RJ, Yorio T, Clark AF, Agarwal N: Characterization of a transformed rat retinal ganglion cell line.
Brain Res Mol Brain Res
86
:
1
–12,
2001
28.
Yu CR, Mahdi RM, Ebong S, Vistica BP, Gery I, Egwuagu CE: Suppressor of cytokine signaling 3 regulates proliferation and activation of T-helper cells.
J Biol Chem
278
:
29752
–29759,
2003
29.
Emanuelli B, Glondu M, Filloux C, Peraldi P, Van Obberghen E: The potential role of SOCS-3 in the interleukin-1beta-induced desensitization of insulin signaling in pancreatic β-cells.
Diabetes
53
(Suppl. 3):
S97
–S103,
2004
30.
Li L, Gronning LM, Anderson PO, Li S, Edvardsen K, Johnston J, Kioussis D, Shepherd PR, Wang P: Insulin induces SOCS-6 expression and its binding to the p85 monomer of phosphoinositide 3-kinase, resulting in improvement in glucose metabolism.
J Biol Chem
279
:
34107
–34114,
2004
31.
Reiter CE, Sandirasegarane L, Wolpert EB, Klinger M, Simpson IA, Barber AJ, Antonetti DA, Kester M, Gardner TW: Characterization of insulin signaling in rat retina in vivo and ex vivo.
Am J Physiol Endocrinol Metab
285
:
E763
–E774,
2003
32.
Hao W, Wenzel A, Obin MS, Chen CK, Brill E, Krasnoperova NV, Eversole-Cire P, Kleyner Y, Taylor A, Simon MI, Grimm C, Reme CE, Lem J: Evidence for two apoptotic pathways in light-induced retinal degeneration.
Nat Genet
32
:
254
–260,
2002
33.
Dudley AC, Thomas D, Best J, Jenkins A: A VEGF/JAK2/STAT5 axis may partially mediate endothelial cell tolerance to hypoxia.
Biochem J
390
:
427
–436,
2005
34.
Chen H, Ye D, Xie X, Chen B, Lu W: VEGF, VEGFRs expressions and activated STATs in ovarian epithelial carcinoma.
Gynecol Oncol
94
:
630
–635,
2004
35.
Gray MJ, Zhang J, Ellis LM, Semenza GL, Evans DB, Watowich SS, Gallick GE: HIF-1alpha, STAT3, CBP/p300 and Ref-1/APE are components of a transcriptional complex that regulates Src-dependent hypoxia-induced expression of VEGF in pancreatic and prostate carcinomas.
Oncogene
24
:
3110
–3120,
2005
36.
Xu Q, Briggs J, Park S, Niu G, Kortylewski M, Zhang S, Gritsko T, Turkson J, Kay H, Semenza GL, Cheng JQ, Jove R, Yu H: Targeting Stat3 blocks both HIF-1 and VEGF expression induced by multiple oncogenic growth signaling pathways.
Oncogene
24
:
5552
–5560,
2005
37.
Barthel A, Schmoll D, Unterman TG: FoxO proteins in insulin action and metabolism: trends in endocrinology and metabolism.
Trends Endocrinol Metab
16
:
183
–189,
2005
38.
Egwuagu CE, Mahdi RM, Chan CC, Sztein J, Li W, Smith JA, Chepelinsky AB: Expression of interferon-gamma in the lens exacerbates anterior uveitis and induces retinal degenerative changes in transgenic Lewis rats.
Clin Immunol
91
:
196
–205,
1999
39.
Egwuagu CE, Sztein J, Mahdi RM, Li W, Chao-Chan C, Smith JA, Charukamnoetkanok P, Chepelinsky AB: IFN-gamma increases the severity and accelerates the onset of experimental autoimmune uveitis in transgenic rats.
J Immunol
162
:
510
–517,
1999
40.
Howard JK, Flier JS: Attenuation of leptin and insulin signaling by SOCS proteins.
Trends Endocrinol Metab
17
:
365
–371,
2006
41.
Ueki K, Kondo T, Tseng YH, Kahn CR: Central role of suppressors of cytokine signaling proteins in hepatic steatosis, insulin resistance, and the metabolic syndrome in the mouse.
Proc Natl Acad Sci U S A
101
:
10422
–10427,
2004
42.
Nishijima K, Ng YS, Zhong L, Bradley J, Schubert W, Jo N, Akita J, Samuelsson SJ, Robinson GS, Adamis AP, Shima DT: Vascular endothelial growth factor-A is a survival factor for retinal neurons and a critical neuroprotectant during the adaptive response to ischemic injury.
Am J Pathol
171
:
53
–67,
2007
43.
Kilic U, Kilic E, Jarve A, Guo Z, Spudich A, Bieber K, Barzena U, Bassetti CL, Marti HH, Hermann DM: Human vascular endothelial growth factor protects axotomized retinal ganglion cells in vivo by activating ERK-1/2 and Akt pathways.
J Neurosci
26
:
12439
–12446,
2006
44.
Hilfiker-Kleiner D, Limbourg A, Drexler H: STAT3-mediated activation of myocardial capillary growth.
Trends Cardio Med
15
:
152
–157,
2005
45.
Leal EC, Santiago AR, Ambrosio AF: Old and new drug targets in diabetic retinopathy: from biochemical changes to inflammation and neurodegeneration.
Curr Drug Targets CNS Neurol Disord
4
:
421
–434,
2005
46.
Mechoulam H, Pierce EA: Expression and activation of STAT3 in ischemia-induced retinopathy.
Invest Ophthalmol Vis Sci
46
:
4409
–4416,
2005
47.
Lu Y, Fukuyama S, Yoshida R, Kobayashi T, Saeki K, Shiraishi H, Yoshimura A, Takaesu G: Loss of SOCS3 gene expression converts STAT3 function from anti-apoptotic to pro-apoptotic.
J Biol Chem
281
:
36683
–36690,
2006

Supplementary data