Investigative Ophthalmology & Visual Science Cover Image for Volume 49, Issue 7
July 2008
Volume 49, Issue 7
Free
Physiology and Pharmacology  |   July 2008
Effect of Intravitreal Administration of Somatostatin and sst2 Analogs on AMPA-Induced Neurotoxicity in Rat Retina
Author Affiliations
  • Foteini Kiagiadaki
    From the Laboratory of Pharmacology, Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece.
  • Kyriaki Thermos
    From the Laboratory of Pharmacology, Department of Basic Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece.
Investigative Ophthalmology & Visual Science July 2008, Vol.49, 3080-3089. doi:https://doi.org/10.1167/iovs.07-1644
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Foteini Kiagiadaki, Kyriaki Thermos; Effect of Intravitreal Administration of Somatostatin and sst2 Analogs on AMPA-Induced Neurotoxicity in Rat Retina. Invest. Ophthalmol. Vis. Sci. 2008;49(7):3080-3089. https://doi.org/10.1167/iovs.07-1644.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

purpose. The aim of the present study was to use an in vivo model of retinal excitotoxicity to investigate the neuroprotective effect of somatostatin (SRIF)-ergic agents.

methods. Adult Sprague–Dawley rats (weight range, 250–300 g) intravitreally received (RS)-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid hydrobromide (AMPA; 21, 42, 84 nmol/eye) or PBS (50 mM). Time-dependent responses were examined in animals that received AMPA (42 nmol/eye). Animals received AMPA (42 nmol) alone or in combination with SRIF (10−5, 10−4 M) or the sst-selective ligands lanreotide (sst2,10−5,10−4 M), L-779976 (sst2,10−6,10−5, 10−4 M), L-797591 (sst1,10−4 M), and L-803087 (sst4,10−4 M). Immunohistochemistry and TUNEL studies were used to examine retinal cell loss and protection. Immunochemistry, Western blot analysis, and radioimmunoassay assessed the viability of sst2A receptors and SRIF levels, respectively, in control and AMPA-treated tissue.

results. AMPA (42 nmol) treatment resulted in total and major loss of ChAT and bNOS immunoreactivity, respectively, 24 hours after its administration. This loss was sustained up to 30 days for ChAT- and 8 days for bNOS-expressing amacrine cells. SRIF and the sst2 receptors were not affected by AMPA. SRIF and the sst2 analogs protected the retina from the AMPA insult in a dose-dependent manner, whereas activation of the sst1 and sst4 subtypes had no effect. TUNEL staining confirmed AMPA-induced retinal ischemia and L-779976 neuroprotection.

conclusions. These results demonstrate for the first time that SRIF and the sst2 analogs, administered intravitreally, protect the retina from excitotoxicity. Further studies are essential to ascertain the therapeutic relevance of these results.

Ischemia is the underlying cause of many ocular diseases that lead to blindness. A large volume of evidence has implicated glutamate excitotoxicity as a leading player in retinal ischemia. 1 Glutamate is the leading neurotransmitter in the retina. It is released from photoreceptors, bipolar cells, and ganglion cells in the vertical signal pathway and plays a major role in vision transduction by activating different receptor subtypes belonging to the ionotropic (N-methyl-d-aspartate [NMDA], kainite, and (RS)-α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid hydrobromide [AMPA] receptors) and metabotropic receptor families. 2 3  
Overstimulation of glutamate receptors leads to retinal toxicity. The NMDA receptor family has been widely studied, and evidence from in vitro and in vivo studies has suggested a leading role for NMDA receptors in retinal excitotoxicity. 1 4 5 Intravitreal injection of NMDA into the adult rat eye caused primarily ganglion but also amacrine cell loss, whereas agents that act as antagonists have protected these retinal neurons. 6 7 It is accepted that the activation of NMDA receptors, the increase in calcium ions, and the subsequent activation of nitric oxide radical formation is the underlying mechanism involved in excitotoxicity. 1 The non-NMDA receptors kainate and AMPA have also been implicated in retinal ischemic insult. 8 9 10 Kainate induced cultured chick amacrine-like cell damage, and this was shown to be mediated by AMPA. 11 Intraocular injections of kainate caused changes in GABA and choline acetyltransferase (ChAT)-containing cells, 12 13 whereas kainate/AMPA receptor blockade was effective in retinal neuroprotection. 1 14  
Many strategies have been used to develop therapeutic agents for the successful treatment of ischemia-induced retinopathies and the prevention of blindness. However, the lack of success so far in attaining this goal makes more evident the complexity of the systems involved in the pathophysiology of these diseases and, therefore, their therapeutics. Recently, we focused on the use of the neuropeptide SRIF as a putative neuroprotective agent in retinal ischemia. 
SRIF 15 is found in the retina, 16 where it activates SRIF receptors, 17 ssts, found in retinal neurons and the retinal pigment epithelium (RPE; for a review, see Thermos 18 ). Specifically, sst2A receptors are localized in rod bipolar, amacrine, and photoreceptor neurons in the retina, 19 20 21 22 23 24 and sst2B receptors are localized in photoreceptors and the RPE. 22 24 In addition, sst1 and sst4 receptors are present in amacrine (SRIF and TH-containing) 25 and ganglion cells, 26 27 respectively. 
SRIF and its analog, sst2, inhibit ischemia-induced neovascularization in a mouse model of oxygen-induced retinopathy 28 29 and depict neuroprotective effects against different paradigms of neurotoxicity in the central nervous system, such as NMDA, kainate-induced neurotoxicity, 30 31 and middle cerebral artery. 32 We used an in vitro model 33 of chemical ischemia in the retina to assess the neuroprotective actions of SRIF. 34 Chemical ischemia involves the blockade of oxidative phosphorylation and glycolysis and is believed to be useful in the understanding of the early events underlying the pathophysiology of ischemia. In this model, SRIF analogs selective for the sst2 subtype protected the retina from the ischemic insult. 34 In a subsequent study using transgenic mice, Catalani et al. 35 showed that increased levels of the sst2 subtype protected the retina from ischemia (hypoxia and iodoacetic acid). This study is in agreement with our previous data. 34 It indirectly showed the involvement of the sst2 subtype and recommended that sst2 receptor agonists may be useful in retinal diseases. 
The aim of the present investigation was to examine SRIF actions in vivo. To this end, an in vivo model of excitotoxicity was used, and the assessment of the ability of SRIF and sst2 analogs to protect the retina, when administered intravitreally, was attempted. AMPA was chosen as the excitotoxic agent, and initial studies focused on the experimental conditions necessary for AMPA-induced retinal cell loss. 
Methods
Animals
Adult male and female Sprague–Dawley rats (weight range, 250–350 g) were housed at room temperature two to three animals per cage and had free access to food and water. A 12-hour light/12-hour dark cycle was maintained. Euthanatization was performed with ether inhalation. All procedures were conducted in accordance with the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research and in compliance with Greek national laws (Animal Act, P.D. 160/91). 
AMPA Excitotoxicity and Neuroprotection
The animals were anesthetized with intramuscular injections of xylazine (14.5 mg/kg) and ketamine (200 mg/kg) and were placed in a stereotaxic instrument to stabilize the head and facilitate intravitreous administration. Injections were made with 27- or 30-gauge needles connected to a Hamilton syringe adapted to a minipump. The tip of the needle was inserted behind the sclera–cornea border into the vitreous humor. Each intravitreal injection was performed with a flow rate of 1 μL/min for 5 minutes. 
One eye received phosphate-buffered saline (50 mM K2HPO4/NaH2PO4, 0.9% NaCl, pH 7.4) as control, and the other eye received AMPA (Tocris) 21, 42, or 84 nmol/eye, respectively, diluted in PBS 50 mM. Rats administered AMPA (42 nmol/eye) were humanely killed at different times (1, 2, 4, 8, 16, and 30 days) after treatment. Neuroprotection experiments involved the injection of AMPA (42 nmol/eye) alone and in combination with SRIF (10−5, 10−4M) or the sst-selective ligand lanreotide (sst2 10−5, 10−4 M), L-779976 (sst2 10−6, 10−5, 10−4 M), L-797591 (sst1 10−4 M), or L-803087 (sst4 10−4 M). Control retinas received PBS (50 mM). 
Immunohistochemical Studies
Tissue Preparation.
After each treatment, the eyes were removed and the eyecups were fixed by immersion in 4% paraformaldehyde in 0.1 M phosphate buffer for 1 hour at 4°C. After fixation, eyecups were rinsed in phosphate buffer and were incubated in 30% sucrose overnight at 4°C for cryoprotection. Tissues were frozen in isopentane at −45°C for 1 minute and kept at −80°C until further use. Eyecups were sectioned vertically at 10-μm thickness using a cryostat, thaw mounted on slides (Superfrost; Erie Scientific, Portsmouth, NH), and stored at −20°C. Slices were cut near the optic nerve every 100 μm. Nine slices were put on every slide. 
Immunohistochemistry.
A mouse monoclonal antibody raised against ChAT (1:100; Biotrend, Cologne, Germany) and a rabbit polyclonal antibody raised against bNOS (1:1000; Sigma, St. Louis, MO) were used as markers for, respectively, acetylcholine and neuronal nitric oxide synthase containing retinal cells. Mouse monoclonal antibodies for protein kinase C (PKC; 1:50; Leinco Technologies, St. Louis, MO) and for microtubule associating protein (MAP)-1 (1:100; Sigma) were used as markers for rod bipolar cells and ganglion cells, respectively. Rabbit polyclonal antibodies for tyrosine hydroxylase (TH) (1:1000; Chemicon, Temecula, CA) and for recoverin (0.044 mg/mL; gift from Eleonora N. Grigoryan) were used as markers for dopamine-containing amacrine cells and photoreceptors/cone bipolar cells, respectively. A mouse monoclonal antibody raised against SRIF (2 μg/mL; Abcam, Cambridge, UK) and a rabbit polyclonal antibody for the SRIF receptor subtype sst2A (1:1000; Gramsch Laboratories, Schwabhausen, Germany) were also used for the detection of SRIFergic- and sst2A-expressing retinal cells, respectively. 
Cryostat sections were incubated in 0.1 M Tris-HCl buffer, pH 7.4, containing 3.3% normal goat serum for 30 minutes, washed in 0.1 M Tris-buffered saline (TBS), and incubated with the primary antibodies in 0.1 M TBS containing 0.3% Triton X-100 and 0.5% normal goat serum overnight at room temperature. The sections were washed again and incubated for 1 hour with the appropriate secondary antibody, Alexa-Fluor 488 goat anti-mouse IgG (H+L; 1:400; Molecular Probes, Eugene, OR) for the monoclonal antibodies or Alexa-Fluor 546 goat anti-rabbit IgG (H+L; 1:400; Molecular Probes) for the polyclonal antibodies. Finally, the sections were washed and coverslipped with mounting medium with DAPI (Vector Laboratories, Burlingame, CA). Negative controls were included by omitting the primary antibody. 
TUNEL Staining.
To determine cell loss, enzymatic in situ labeling of apoptosis-induced DNA strand breaks was performed on tissue sections obtained from retinas 1 day after AMPA treatment. The terminal deoxynucleotidyl transferase (TDT)-mediated TMR-dUTP nick-end labeling (TUNEL) assay (Roche, Germany) was used. 
Microscopy.
Light microscopy images were taken with a camera (Axioskop with Plan-Neofluar ×40/0.75; Carl Zeiss, Oberkochen, Germany). Optic sections were taken with a z-axis resolution of 1.1 μm through the immunolabeled cells. Light and contrast adjustments of images were processed with the use of commercial software (Photoshop ver. 7.0; Adobe Systems, San Jose, CA). 
Quantification
For the quantification of bNOS-expressing retinal cells, each section of control or treated retina was studied by means of a ×40/0 lens (Planapo; Hewlett-Packard, Palo Alto, CA). The total number of bNOS-immunoreactive neurons in each retinal section was counted. As mentioned in Tissue Preparation, for the immunohistochemistry studies, all slices used were cut near the optic nerve. Nine slices were put on every slide. bNOS-expressing cells were counted on all nine sections per slide. Therefore, the maximum number of sections that could be examined was n (number of retinas) × 9. Given that some slices were not appropriate for cell counting (e.g., because of tissue folding or damage during cutting), the actual number of sections used for the quantification was equal to or less than n × 9. 
The numbers of sections used were as follows: control groups: 1-day group, 41 sections were used from 5 different retinas (n = 5); 8-day group, 20 sections were used from 3 different retinas (n = 3), 16-day group, 27 sections were used from 3 different retinas (n = 3), 30-day group, 27 sections were used from 3 different retinas (n = 3); AMPA-treated groups: 1-day group, 26 sections were used from 4 different retinas (n = 4); 8-day group, 23 sections were used from 3 different retinas (n = 3); 16-day group, 20 sections were used from 3 different retinas (n = 3); 30-day group, 25 sections were used from 3 different retinas (n = 3). Twenty-seven sections from 3 different retinas (n = 3) were used for the AMPA+L-779976–treated group (neuroprotection studies). 
Radioimmunoassay
SRIF levels were examined in retinas that were removed from eyes treated with PBS (50 mM; n = 6) or AMPA (42 nmol/eye; n = 7) 1 day after treatment. Samples were prepared according to the method described by Mastrodimou and Thermos. 36 In short, retinas were centrifuged for 15 minutes at 12,000 rpm and 4°C. Acetic acid (2 N) was added to the precipitant, and the mixture was boiled for 10 minutes, homogenized with a sonicator, and stored at −80°C for 24 to 48 hours. Subsequently, the mixture was centrifuged for 20 minutes at 13,000 rpm, and the supernatant was lyophilized and kept at −80°C. The assay mixture contained the SRIF antiserum (kindly provided by Günther Sperk) at a final dilution of 1:3000, [125I]-Tyr 11 -SRIF (20,000 cpm), and SRIF standards (0–400 pg/100 μL) or samples in a final assay volume of 0.5 mL, according to Sperk and Windmann. 37 Incubation was carried out at 4°C for 24 hours. Separation of bound and free peptide was achieved by the addition of 900-μL aliquots of a mixture containing 2.5% charcoal and 0.25% dextran T70 and subsequent centrifugation. 
Western Blotting
Retinal Membrane Preparation.
Sprague–Dawley rats were anesthetized, and retinas were removed and homogenized (Ultra-Turrax homogenizer; IKA Works, Staufen, Germany) in Tris-HCl, pH 7.4. The homogenate was centrifuged at 1000g for 10 minutes, and the supernatant was aspirated and stored. The pellet was resuspended, rehomogenized, and centrifuged as described. The two supernatants were combined and centrifuged at 11,000g for 20 minutes. The pellet was resuspended in Tris buffer and centrifuged at 27,000g for 10 minutes. Finally, the pellet was resuspended in lysis buffer (10 mM Tris-HCl, pH 7.4, 250 mM sucrose, 1 mM EDTA, 1 mM phenylmethylsulfonyl fluoride, 1% Triton, 2 μg/mL leupeptin, 2 μg/mL aprotinin). All steps of this procedure were performed at 4°C. The protein content was determined according to Bradford, 38 and aliquots of each sample containing equal amounts of protein (32 μg) were subjected to 10% SDS-PAGE and immunoblotted onto nitrocellulose membranes. Blots were incubated with anti-sst2A (1:2500) overnight at 4°C. For adsorption controls, sst2A antigen at concentrations of 10−4 M were incubated with the antisera for 2 hours at room temperature. Blots were developed using peroxidase-conjugated secondary antibodies and enhanced chemiluminescence. These experiments were performed two to four times from the same or different preparations of retinas. 
Retinal Protein Samples from Treated Animals.
Animals were humanely killed, and retinas were removed and homogenized (Ultra-Turrax homogenizer; IKA Works) in lysis buffer. Because of the scarcity of the tissues, the whole membrane preparation was used. Retinal samples (60 μg) were subjected to 10% SDS-PAGE and immunoblotted onto nitrocellulose membranes. Blots were incubated with a rabbit polyclonal antibody against SRIF receptor subtype sst2A (1:2500; Gramsch Laboratories) overnight at 4°C. Blots were developed using peroxidase-conjugated secondary antibodies and enhanced chemiluminescence. These experiments were performed two to four times using protein samples from the same or different preparations of retinas. 
Statistical Analysis
Analysis of data was performed using a one-way ANOVA with post hoc analysis (Dunnett; GraphPad Prism 2.01; GraphPad Software, San Diego, CA). 
Results
Effect of AMPA on ChAT Immunoreactivity: Dose- and Time-Response Experiments
ChAT immunoreactivity was observed in amacrine cells located in the inner nuclear layer (INL) and the ganglion cell layer (GCL) and in processes in the inner plexiform layer of PBS-treated retinas (Fig. 1A) . Intravitreal administration of the excitatory amino acid AMPA (21, 42, 84 nmol/eye) caused a dose-dependent loss of ChAT expression. The intravitreal injection of AMPA (21 nmol/eye) caused a partial loss of ChAT immunoreactivity because processes were still visible 1 day after injection. AMPA (42 nmol/eye) caused a complete loss of ChAT expression, whereas a higher concentration of 84 nmol/eye gave similar results (Fig. 1) . ChAT immunoreactivity loss was sustained for 8, 16, or 30 days after AMPA administration (Fig. 2) . The effect of AMPA on ChAT immunoreactivity was also examined 2 and 4 days after injection, and similar results were obtained (data not shown). 
Effect of AMPA on PKC, TH, bNOS, Recoverin, and MAP1A Immunoreactivity
To examine the effect of the AMPA treatment (42 nmol/eye) on other retinal cell types, immunoreactivity studies were performed using antibodies against retinal markers that recognize photoreceptors and rod and cone bipolar, amacrine, and ganglion cells. Figure 3depicts the PKC and MAP1A immunoreactivities in retinal tissues isolated 1 day after AMPA treatment. PKC immunoreactivity was not different in the two groups of retinas (n = 2), even though the staining pattern in Figure 3Bmight have suggested more dense staining in rod bipolar cells of the AMPA-treated retinas. Similarly, MAP1A immunoreactivity was rich in the processes and some cell bodies of ganglion cells (Figs. 3C 3D) , but no differences were observed between control and AMPA-treated retinas (n = 3). TH immunoreactivity in amacrine cells was not affected by the AMPA treatment (n = 3; Figs. 4A 4B ). Recoverin immunoreactivity was evident in the outer nuclear layer (ONL) and the INL in control and AMPA-treated retinas (n = 2; Figs. 4C 4D ). Immunohistochemical studies using the same antibodies were performed on tissues obtained 8, 16, and 30 days after AMPA treatment. Results were similar to those described for the 1-day groups (data not shown). 
bNOS immunoreactivity studies showed that NOS-containing neurons were decreased 1 day after AMPA administration. However, the signal returned to control levels 8 days after AMPA injection. To avoid the repetition of immunoreactivity staining patterns, bNOS-containing amacrine cells were counted in PBS- and AMPA-treated retinal sections, and the results are shown in Figure 5
Effect of AMPA on SRIF and sst2A Levels
The most significant changes in ChAT and bNOS immunoreactivities were observed as early as 1 day after AMPA (42 nmol/eye) treatment. Therefore, we examined whether SRIF levels and the expression of the sst2 receptor, the most predominant receptor subtype in the retina, were affected in the same paradigm. SRIF levels were measured by radioimmunoassay. No statistically significant changes were observed between the control and the AMPA-treated retinas (Fig. 6) . The presence of the sst2 receptor in the retina was examined by immunoreactivity and Western blot methodologies. Immunoreactivity differences with sst2A were not apparent between the PBS- and AMPA-treated retinas (Figs. 7A 7B) . To analyze further the retinal samples under study, Western blot analysis was performed. Initially, we performed control studies to examine the presence of the receptor in retinal membranes. A protein (molecular mass, approximately 77 kDa) was detected in the retina that was not visible in the presence of the sst2 antigen (Fig. 7C) . Therefore, we concluded that the 77-kDa protein represented the sst2 receptor subtype. 
Western blot analysis using crude protein preparations of PBS- and AMPA-treated retinas depicted a protein of apparent molecular weight of approximately 72 kDa (Fig. 7D) . We presumed this protein to be the equivalent of the 77-kDa protein in the control blots. Actin immunoblots were also examined in the two samples to substantiate that equal protein concentrations were applied, and no differences were evident between the PBS- and AMPA-treated retinas. 
Effect of Somatostatin on AMPA-Induced Loss of ChAT Immunoreactivity
To examine whether SRIF could protect the retina from the AMPA-induced loss of ChAT immunoreactivity, SRIF was coinfused with AMPA (42 nmol/eye). ChAT immunoreactivity was assayed 1 day after the treatment. SRIF protected the cholinergic neurons in a dose-dependent manner (10−5, 10−4 M; Fig. 8 ), but it did not afford full protection. 
Effect of the Selective Agonists Lanreotide (sst2/5) and L-779976 (sst2) on AMPA-Induced Loss of ChAT and bNOS Immunoreactivity
The putative neuroprotective action of selective analogs of SRIF receptors was also studied (Fig. 9) . Lanreotide (10−5 M) coinfused with AMPA (42 nmol/eye) mimicked the effects of SRIF at 10−4 M (Fig. 9C) . ChAT immunoreactivity completely recovered at the higher lanreotide concentration of 10−4 M (Fig. 9D) . The sst2-selective ligand L-779976 was also examined. As shown in Figure 10 , L-779976 protected the retina in a dose-dependent manner, with full recovery at the highest dose used (10−4M). bNOS immunoreactivity was also examined in retinas treated with L-779976 (10−4 M). The viable cells observed in the presence of L-779976 were comparable to the control samples, suggesting full recovery (Fig. 10F)
TUNEL staining was used to assess retinal cell death by AMPA (42 nmol/eye) and protection by the L-779976 analog 1 day after treatment. The results, shown in Figure 11 , confirm the excitotoxic effect of AMPA and the protection afforded by L-779976 (10−4 M). 
Effect of the sst1- and sst4-Selective Analogs on AMPA-Induced Loss of ChAT Immunoreactivity
Receptor subtypes sst1 and sst4 have also been detected in amacrine and ganglion cells, respectively, of rat retina. 26 Therefore, the putative neuroprotective effect of selective sst1 and sst4 analogs was examined. The AMPA-induced loss of ChAT immunoreactivity was not reversed by the presence of L-797591 (sst1 selective) or L-803087 (sst4 selective) at the concentration of 10−4 M, a concentration that was effective for the sst2 ligands (Fig. 12)
Discussion
The main finding of this study is that SRIF and its analogs, lanreotide and L-779976, provide neuroprotection against excitatory insults in vivo. Injected intravitreally, these agents reduce the retinal damage induced by the excitatory amino acid AMPA. 
Glutamate, the major neurotransmitter in the retina, is responsible for the transfer of visual information from the retina to the brain. It is released by photoreceptors, bipolar cells, and ganglion cells and acts on ionotropic and metabotropic glutamate receptors found in retinal neurons. 2 39 40 Excess glutamate release and overactivation of its ionotropic receptors is believed to be associated with toxicity and cell death. Therefore, the use of glutamate and other excitatory amino acids with high affinity for the ionotropic receptor family, such as NMDA, kainate, and AMPA, have been used as models of excitotoxicity. These studies have been instrumental in the evaluation of the events leading to toxicity caused by ischemic insults because excitotoxicity is believed to be responsible for the ischemia-related cell death. 1  
In the present study, we used AMPA and examined its effects when given intravitreally in the retina. Subsequently, we studied whether the neuropeptide SRIF could reverse the toxic insult. AMPA receptors belong to the non-NMDA family of ionotropic glutamate receptors. They are composed of four subunits, GluR1 to GluR4, that are differentially expressed in retina. 41 42 AMPA receptors lacking GluR2 have an increased permeability to intracellular calcium ions, making the neurons that express these receptors more vulnerable. 3 43 44  
The present data showed that AMPA differentially influenced retinal cell viability. It eliminated, in a dose-dependent manner, ChAT immunoreactivity 24 hours after its injection into the rat eye. This loss persisted 30 days after the AMPA intravitreal injection. Similarly, it reduced the number of bNOS-containing amacrine cells. However, the signal recovered 8 days after injection, in contrast to what was observed for the ChAT immunoreactivity signal. No changes were observed in the retinal markers used to label photoreceptors or cone and rod bipolar, TH-containing amacrine and ganglion cells. 
The degree of loss observed in the different cell types is directly coupled to the presence of the AMPA receptor subtype. mRNA and protein studies support the localization of AMPA receptors in all retinal cell types of different animal species, 2 40 45 but a differential localization of the AMPA receptor subunits has been reported. 45 In agreement with the present findings, ChAT immunoreactivity was shown to be obliterated after intravitreal kainic 13 and quisqualic acid injection. 46 The presence and function of AMPA receptors on cholinergic neurons was also shown by release studies. Selective blockade of AMPA receptors in rabbit retina inhibited the light-evoked increase in acetylcholine release, whereas the subunits GluR2 and GluR3 were found to be localized on ChAT immunoreactive processes. 47 A drastic reduction in ChAT immunoreactivity was also observed after exposure of the retinas to kainate and in a model of high intraocular pressure. The authors suggested that in paradigms in which the ischemia is of sufficient severity, kainate receptors associated with cholinergic neurons are overstimulated, leading to membrane disruption and enzyme (ChAT) release. 13 This may reflect the changes observed in the present paradigm in which AMPA receptors are activated, leading to ChAT immunoreactivity loss. 
An earlier study, examining the effect of excitotoxicity in the rabbit retina, indicated that NADPH-diaphorase (marker for NOS) amacrine cells contained NMDA and kainic acid receptors. 48 In addition, in turtle retina, NOS inhibitors antagonized the kainic acid increase in cGMP-like immunoreactivity, suggesting that the activation of kainic acid receptors in NOS-containing neurons stimulate NO release. 49 To our knowledge, there are no immunohistochemical data to support the localization of AMPA receptors on NOS-containing amacrine cells in the retina. However, these data pertaining to the use of kainic acid (also an agonist of AMPA receptors) indirectly support the presence of AMPA receptors and their implication in the loss of bNOS immunoreactivity in the present study. In contrast to ChAT immunoreactivity, bNOS immunoreactivity reappeared 8 days after AMPA treatment. The morphology of the bNOS-expressing cells was the same as in the control samples (data not shown). 
In an extensive immunocytochemical study, the differential effects of ischemia/reperfusion on different amacrine cell subtypes was examined. Loss of immunoreactivity was observed in amacrine cells expressing Glyt1 (glycinergic), substance P, and CR (subpopulation of cholinergic cells) at 2 hours after ischemia, but the immunoreactivity recovered to a large extent between 4 and 12 hours after ischemia. ChAT immunoreactivity in this paradigm was reduced with no subsequent recovery. 50 This latter study and the present results support differential changes in amacrine cells as a result of ischemia/reperfusion and excitatory amino acid receptor activation, respectively. The transient loss of immunoreactivity may be the result of the inhibition of protein synthesis and its subsequent restoration, whereas the ChAT immunoreactivity loss may be a result of the stimulation of protease activity (caspases) and long-term effects. 50 Ischemia-reperfusion injury was shown to activate different caspases depending on the neuronal phenotype in the retina, with caspases 2 and 3 acting in parallel in amacrine neurons. 51  
In agreement with the present data, intravitreal infusion of quisqualic acid did not affect photoreceptors, bipolar cells, or ganglion cells, and only a small loss of TH-containing amacrine cells from the central retina was reported. 46 Furthermore, GluR1–4 expression was not found in photoreceptors, 45 suggesting that AMPA administration should have no effect on these retinal neurons. Further evidence in the literature supports the lack of effect of AMPA on rod and cone bipolar and ganglion cells. Electrophysiological studies have shown 2-amino-4-phosphonobutyric acid (APB)-sensitive functional responses on the activation of rod and cone ON-bipolar cells. 52 These bipolar cells are activated by glutamate primarily through the APB-type metabotropic receptor and involve cGMP signaling. 53 However, recent colocalization immunohistochemical studies supported the presence of AMPA-type glutamate receptors (GluR2) in PKC-containing cells (rod and cone ON-bipolar) in the rat retina, 54 whereas single-cell RT-PCR showed the presence of GluR1/GluR2 in rod and cone bipolar cells. 45 As stated, the presence of GluR2 renders the receptor impermeable to calcium ions; thus, its presence in neurons should spare them from toxicity. Kainate and NMDA receptors are expressed in ganglion cells, 55 56 and GluR4 mRNA has also been detected. 45 The relative survival of photoreceptors, bipolar cells, and ganglion cells after AMPA exposure may be attributed to the absence or low levels of the receptor, as mentioned, or to their ability to deal with sustained depolarization and ionic fluctuation. It was suggested that the large cytosolic volume of ganglion cells might buffer the accumulation of intracellular calcium and thus make these cell less vulnerable to excitotoxicity. 46  
The findings thus far suggested that AMPA is a good model of retinal cell loss (subtypes of amacrine cells) and support its use for the testing of agents as neuroprotectants of retinal neurons against toxicity. To this end, the subsequent studies investigated whether SRIF and specific sst analogs could be useful in the reversal of the actions of AMPA. SRIF is released from a subclass of amacrine cells in the retina 16 36 and activates its receptors, sst1 to sst5, found in different retinal neurons. 20 The sst2 subtype appears to be the predominant one in vertebrate retina. 
AMPA did not have any effect on SRIF levels, as measured by radioimmunoassay (Fig. 6) . This suggests that AMPA receptors are not present in SRIF-containing amacrine neurons. In addition, the intravitreal injection of AMPA did not influence the sst2 receptors, as shown by immunohistochemical and Western blot analysis studies (Fig. 7) . The presence of viable sst2 receptors enabled us to proceed with the investigation of whether SRIF itself and the specific sst2 analogs could protect the retina from AMPA excitotoxicity. As shown by ChAT immunoreactivity, intravitreal administration of SRIF was able to partially reverse the damage imposed by AMPA on the retina (Fig. 8) . The lack of full recovery of the signal may be attributed to peptide degradation. However, similar results were obtained in the presence or absence of protease inhibitors (data not shown). Thus, other factors (e.g., absorption) may influence SRIF action. The sst2-selective ligands lanreotide and L-779976 were able to reverse the AMPA damage and bring ChAT immunoreactivity to control levels (Figs. 9 10)
TUNEL staining was in agreement with the ChAT immunoreactivity data, showing increased apoptosis only in the AMPA-treated tissue and providing further support for the neuroprotective actions of the sst2 analog (Fig. 11) . However, TUNEL labeling was observed in all retinal layers. This may suggest that AMPA receptor expression, which has not been detected in some retinal cells to date, may actually exist or that AMPA activation of caspases may be lead to cell death in other retinal cell layers. 57 58 The extensive TUNEL labeling, however, does not correlate with the lack of change of expression in retinal markers other that ChAT. This is in partial agreement with a recent study in the mouse that used iodoacetic acid and hypoxia to develop a retinal ischemia model. 35 TUNEL labeling was observed in the three retinal layers—ONL, INL, and GCL—1 hour after exposure of the retina to the ischemia mixture, yet markers such as cone arrestin (marker for cone photoreceptors) were not affected up to 6 hours (maximum exposure) of ischemic treatment. In the same study, it was observed that different retinal markers were differentially affected according to the time of exposure to the ischemia mixture. ChAT immunoreactivity was completely lost as early as 1 hour and remained low, TH immunoreactivity was decreased but was still evident after 6 hours of ischemia, and TUNEL labeling increased as a function of time. 35 In a model of chemical ischemia (iodoacetic acid and sodium cyanide), rod bipolar, ChAT, TH, and bNOS-expressing amacrine cells were mostly affected. However, TUNEL labeling was observed predominantly in the ONL and INL layers. 34  
Retinal ischemia models involving hypoxia and hypoglycemia lead to the release of glutamate and the activation of NMDA and non-NMDA receptors and to subsequent toxicity and cell death. 1 34 35 A lack of concordance between TUNEL labeling and retinal marker expression was also observed in a study that characterized quisqualic acid and NMDA-induced excitotoxicity in chick retina. In this study, DNA fragmentation was observed in rod bipolar cells in which no detectable loss of PKC immunoreactivity was detected. The authors suggested that some cells damaged by the excitatory amino acid, though labeled by the TUNEL technique, were either not committed to apoptosis or were rescued by DNA repair. 46 The lack of concordance between TUNEL labeling and retinal marker proteins and between protein and retinal marker transcripts 59 has been observed in more paradigms than can be cited in this study. Future investigations focusing on the elucidation of the mechanisms involved in these processes will be important in providing specific answers to the questions posed. 
The present data support that the neuroprotective effect of SRIF is mediated by its activation of sst2 receptors, because sst1- and sst4-selective ligands had no effect on drug concentrations that afforded protection by lanreotide and L-779976 (Fig. 12) . These results are in agreement with previous data from our laboratory that showed sst2-selective ligands to be neuroprotective in a chemical model of retinal ischemia. 34 Octreotide (sst2/5 ligand) was also shown to protect the guinea pig retina from ischemia reperfusion injury. 60 A more recent study 35 complemented these data by showing that the overexpression of sst2 receptors in mice lacking the sst1 subtype prevented the retinal ischemia produced by iodoacetic acid and hypoxia. 
The mechanisms by which SRIFergic ligands act as neuroprotectants are still under investigation. SRIF is known to inhibit voltage-gated calcium channels 61 and neuronal calcium currents, the latter through a mechanism involving a cGMP-dependent protein kinase. 62 cGMP was also important in SRIF-protective actions against NMDA-induced neuronal death in cortical cultures. 30 In a recent study, we showed that NO/peroxynitrite and cGMP are important mediators in the protection of rat retina from chemical ischemia and that a NO/sGC/cGMP signaling pathway is involved in the neuroprotective effects bestowed on the retina by the sst2 SRIF ligands in the same model. 63 Studies are in progress to examine whether such a mechanism is involved in SRIF protection of the retina against the AMPA insult. 
The importance of the present data lies in the fact that SRIF, particularly the two ligands tested (lanreotide and L-779976), afforded neuroprotection against excitotoxicity when administered intravitreally in vivo. Lanreotide is known to have a high affinity for sst2 and sst5 receptors. 64 Until recently, the sst5 subtype was not localized in the retina; thus, we assumed that the neuroprotective effects of lanreotide were restricted to its activation of the sst2 subtype. Nevertheless, a recent study supported the presence of the sst5 receptor in dopaminergic and cholinergic amacrine cells, 65 and the possible contribution of this receptor to lanreotide’s effects cannot be excluded. L-779976 is a selective sst2 ligand. 66 However, one may criticize the concentrations of lanreotide and L-779976 (10−5M, 10−4 M) that were shown to be efficacious as neuroprotectants as high and possibly nonspecific. Similar concentrations were shown to be efficacious in the chemical ischemia model 34 and in an experimental paradigm of choroidal neovascularization. 67 Although these cited data do not justify the protocol, we can support the present findings for two reasons. First, we cannot conjecture about the actual drug concentration reaching the retina (possibly small) because we have no means of measuring drug levels after intravitreal injection. Second, the lack of neuroprotective effects of the sst1- and sst4-selective ligands 66 and the absence of the sst3 receptor in vertebrate retinas 18 suggest that lanreotide and L-779976 mediate their neuroprotective effects through activation of the sst2 and possibly the sst5 receptors. 
Many investigations in the literature have focused on the study of the use of SRIF analogs in the therapeutics of retinal diseases. Proliferative diabetic retinopathy (PDR) is a major cause of visual loss. Its pathophysiology involves increased vascularization or angiogenesis. SRIF has been shown to have antiangiogenic properties 28 29 and possibly to be useful in the treatment of PDR. A recent study reported that SRIF-28 is attenuated in the vitreous fluid of patients with PDR, adding further support to the significance of SRIF analogs in the treatment of this disease. 68 A similar deficit of SRIF was detected in the vitreous of patients with diabetic macular edema. 69 SRIF analogs have been used in a small number of ocular clinical studies. Recently, in a case study, octreotide (100 μg) was administered subcutaneously three times a day for 8 months to a 52-year-old woman with cystoid macular edema (CME). 70 This treatment resulted in improvement of the patient’s visual acuity and partial resolution of CME. In addition, lanreotide (Somatuline; Ipsen, Paris, France) was administered subcutaneously twice a month for 6 months to patients with age-related maculopathy and was found to stabilize the patients’ visual acuity. 71  
The present in vivo data support the pharmacologic and possible therapeutic significance of the intravitreal administration of lanreotide and L-779976. Further studies are essential to examine whether the intravitreal injection of these analogs is devoid of pharmacokinetic problems associated with systemic administration, such as proper drug absorption and distribution. Octreotide acetate was injected into the vitreous cavity of kitten eyes, the long-acting octreotide-LAR was injected into the mid-vitreous of rabbit eyes, and the pharmacokinetics and toxicity of the ligands were examined. 72 This study concluded that intravitreal doses up to 1.1 mg octreotide-LAR could provide sustained high concentrations of the drug that would render it useful as a potential treatment of proliferative eye diseases. Another study evaluated the ocular toxicity of intravitreous octreotide and came to a similar conclusion as to the safety of octreotide at a concentration of less than 1 mg. 73 More recently, a SRIF–camptothecin conjugate was examined for its ocular toxicity and efficacy after intravitreal administration in rabbits. It was found to be safe at the concentration of 10−5 M or less and to be efficacious in the treatment of age-related macular degeneration. 67  
In conclusion, the present study offers new in vivo evidence suggesting that the SRIFergic ligands lanreotide and L-779676 protect the retina from excitatory amino acid damage. The pharmacologic profile of these agents renders them promising therapeutic agents in retinal diseases whose pathophysiologies involve ischemic and excitotoxic insult. There is still a great need for new, effective drugs for the treatment of retinopathies. Therefore, further investigations to substantiate the proper pharmacokinetics and the lack of toxicity of these agents in ocular tissues may render them useful in the treatment of retinopathies. 
 
Figure 1.
 
Dose-dependent effect of AMPA treatment on ChAT immunoreactivity in the retina. ChAT immunoreactivity in control rat retina (A) and in retina that received 21 (B), 42 (C), and 84 (D) nmol AMPA 1 day after injection. Partial loss of ChAT I-R was observed with 21 nmol/eye (only some processes of the cholinergic neurons appear), whereas the larger doses caused the complete loss of ChAT I-R. Scale bar, 50 μm.
Figure 1.
 
Dose-dependent effect of AMPA treatment on ChAT immunoreactivity in the retina. ChAT immunoreactivity in control rat retina (A) and in retina that received 21 (B), 42 (C), and 84 (D) nmol AMPA 1 day after injection. Partial loss of ChAT I-R was observed with 21 nmol/eye (only some processes of the cholinergic neurons appear), whereas the larger doses caused the complete loss of ChAT I-R. Scale bar, 50 μm.
Figure 2.
 
Time-dependent effect of AMPA treatment on ChAT immunoreactivity in the retina. ChAT immunoreactivity in rat retina that intravitreally received PBS (50 mM, control tissue; A) or AMPA (42 nmol/eye) 1 (B), 8 (C), 16 (D), and 30 (E) days after treatment. The complete loss of choline acetyltransferase expression from the first until the thirtieth day after treatment was observed. Arrows: nonspecific binding of the secondary antibody to blood vessels. Scale bar, 50 μm.
Figure 2.
 
Time-dependent effect of AMPA treatment on ChAT immunoreactivity in the retina. ChAT immunoreactivity in rat retina that intravitreally received PBS (50 mM, control tissue; A) or AMPA (42 nmol/eye) 1 (B), 8 (C), 16 (D), and 30 (E) days after treatment. The complete loss of choline acetyltransferase expression from the first until the thirtieth day after treatment was observed. Arrows: nonspecific binding of the secondary antibody to blood vessels. Scale bar, 50 μm.
Figure 3.
 
Effect of AMPA treatment on PKC and MAP expression in the retina. PKC immunoreactivity in control (A) and AMPA (42 nmol/eye)–treated tissue (B), MAP1A immunoreactivity in control (C), and AMPA-treated tissue 1 day after injection (D). Scale bar, 50 μm.
Figure 3.
 
Effect of AMPA treatment on PKC and MAP expression in the retina. PKC immunoreactivity in control (A) and AMPA (42 nmol/eye)–treated tissue (B), MAP1A immunoreactivity in control (C), and AMPA-treated tissue 1 day after injection (D). Scale bar, 50 μm.
Figure 4.
 
Effect of AMPA treatment on TH and recoverin expression in the retina. TH immunoreactivity in control (A) and AMPA (42 nmol/eye)–treated tissue and (B) recoverin immunoreactivity in control (C) and AMPA-treated tissue (D) 1 day after injection. Scale bar, 50 μm.
Figure 4.
 
Effect of AMPA treatment on TH and recoverin expression in the retina. TH immunoreactivity in control (A) and AMPA (42 nmol/eye)–treated tissue and (B) recoverin immunoreactivity in control (C) and AMPA-treated tissue (D) 1 day after injection. Scale bar, 50 μm.
Figure 5.
 
Effect of AMPA on bNOS immunoreactivity. bNOS-containing amacrine cells were counted in PBS and AMPA (42 nmol/eye)–treated retinal sections at 1, 8, 16, and 30 days. NOS-containing neurons were decreased 1 day after AMPA administration. However, the signal returned to control levels 8 days after AMPA injection. Results are presented as mean ± SEM.
Figure 5.
 
Effect of AMPA on bNOS immunoreactivity. bNOS-containing amacrine cells were counted in PBS and AMPA (42 nmol/eye)–treated retinal sections at 1, 8, 16, and 30 days. NOS-containing neurons were decreased 1 day after AMPA administration. However, the signal returned to control levels 8 days after AMPA injection. Results are presented as mean ± SEM.
Figure 6.
 
Effect of AMPA on SRIF levels. SRIF levels were measured by radioimmunoassay. No statistically significant changes were observed between the control and the AMPA (42 nmol/eye)–treated retinas 1 day after injection.
Figure 6.
 
Effect of AMPA on SRIF levels. SRIF levels were measured by radioimmunoassay. No statistically significant changes were observed between the control and the AMPA (42 nmol/eye)–treated retinas 1 day after injection.
Figure 7.
 
Effect of AMPA on sst2A expression levels. sst2A Immunoreactivity in PBS-treated and AMPA (42 nmol/eye)–treated retina 1 day after injection (A, B). sst2A Immunoreactivity is located in somata, axons, and processes of bipolar cells, and no differences between control and excitotoxic tissue were observed. Scale bar, 50 μm. Western blot studies on control retinal membranes (C). A protein of approximate molecular mass of 77 kDa was detected in the retina that was not visible in the presence of the sst2 antigen. No differences in the expression of the receptor protein (72 kDa) were observed in the PBS- and AMPA-treated retinas (D).
Figure 7.
 
Effect of AMPA on sst2A expression levels. sst2A Immunoreactivity in PBS-treated and AMPA (42 nmol/eye)–treated retina 1 day after injection (A, B). sst2A Immunoreactivity is located in somata, axons, and processes of bipolar cells, and no differences between control and excitotoxic tissue were observed. Scale bar, 50 μm. Western blot studies on control retinal membranes (C). A protein of approximate molecular mass of 77 kDa was detected in the retina that was not visible in the presence of the sst2 antigen. No differences in the expression of the receptor protein (72 kDa) were observed in the PBS- and AMPA-treated retinas (D).
Figure 8.
 
Effect of somatostatin on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with SRIF (10−5 M; C,10−4 M; D) 1 day after injection. Partial protection was afforded by SRIF 10−4 M. Scale bar, 50 μm.
Figure 8.
 
Effect of somatostatin on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with SRIF (10−5 M; C,10−4 M; D) 1 day after injection. Partial protection was afforded by SRIF 10−4 M. Scale bar, 50 μm.
Figure 9.
 
Effect of lanreotide (sst2/5 analog) on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with lanreotide (10−5 M; C, 10−4 M; D) 1 day after injection. Lanreotide protected the retina in a dose-dependent manner. Scale bar, 50 μm.
Figure 9.
 
Effect of lanreotide (sst2/5 analog) on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with lanreotide (10−5 M; C, 10−4 M; D) 1 day after injection. Lanreotide protected the retina in a dose-dependent manner. Scale bar, 50 μm.
Figure 10.
 
Effect of L-779976 (sst2 analog) on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with L-779976 (10−6 M; C, 10−5 M; D, 10−4 M; E) 1 day after injection. L-779976 protected the retina in a dose-dependent manner. Scale bar, 50 μm. (F) Effect of AMPA (42 nmol) alone or in combination with L-779976 (10−4 M) on the number of bNOS expressing retinal cells. The AMPA effect is statistically different when compared to the control (*P < 0.05).
Figure 10.
 
Effect of L-779976 (sst2 analog) on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with L-779976 (10−6 M; C, 10−5 M; D, 10−4 M; E) 1 day after injection. L-779976 protected the retina in a dose-dependent manner. Scale bar, 50 μm. (F) Effect of AMPA (42 nmol) alone or in combination with L-779976 (10−4 M) on the number of bNOS expressing retinal cells. The AMPA effect is statistically different when compared to the control (*P < 0.05).
Figure 11.
 
Effect of L-779976 (sst2 analog) on AMPA-induced apoptosis. TUNEL staining used to assess retinal cell death by AMPA and protection by the L-779976 analog 1 day after injection. AMPA afforded excitotoxic effects (B) compared with control (A). L-779976 (10−4 M) protected the retina from AMPA-induced apoptosis (C). Scale bar, 50 μm.
Figure 11.
 
Effect of L-779976 (sst2 analog) on AMPA-induced apoptosis. TUNEL staining used to assess retinal cell death by AMPA and protection by the L-779976 analog 1 day after injection. AMPA afforded excitotoxic effects (B) compared with control (A). L-779976 (10−4 M) protected the retina from AMPA-induced apoptosis (C). Scale bar, 50 μm.
Figure 12.
 
Effect of L-797591 (sst1 analog) and L-803087 (sst4 analog) ligands on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with L-797591 (10−4 M; C) or L-803087 (10−4 M; D) 1 day after injection. Activation of sst1 and sst4 receptors had no effect on the AMPA-induced loss of ChAT expression. Scale bar, 50 μm.
Figure 12.
 
Effect of L-797591 (sst1 analog) and L-803087 (sst4 analog) ligands on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with L-797591 (10−4 M; C) or L-803087 (10−4 M; D) 1 day after injection. Activation of sst1 and sst4 receptors had no effect on the AMPA-induced loss of ChAT expression. Scale bar, 50 μm.
The authors thank Anna Vasilaki and Miltiadis Tsilimbaris for the constructive discussions on this work, Eleni Renieri for excellent technical assistance, and Merck for kindly providing the selective sst L-analogs. 
OsborneNN, CassonRJ, WoodJP, ChidlowG, GrahamM, MelenaJ. Retinal ischemia: mechanisms of damage and potential therapeutic strategies. Prog Retin Eye Res. 2004;23:91–147. [CrossRef] [PubMed]
BrandstätterJH, KoulenP, WässleH. Diversity of glutamate receptors in the mammalian retina. Vis Res. 1998;38:1385–1397. [CrossRef] [PubMed]
LiuSJ, ZukinRS. Ca2+-permeable AMPA receptors in synaptic plasticity and neuronal death. Trends Neurosci. 2007;30:126–134. [CrossRef] [PubMed]
ZeevalkGD, NicklasWJ. Chemically induced hypoglycemia and anoxia: relationship to glutamate receptor-mediated toxicity in retina. J Pharmacol Exp Ther. 1990;253:1285–1292. [PubMed]
SucherNJ, LiptonSA, DreyerEB. Molecular basis of glutamate toxicity in retinal ganglion cells. Vis Res. 1997;37:3483–3493. [CrossRef] [PubMed]
OlneyJ, PriceM, SallesKS, LabruyereJ, FrierdichG. MK-801 powerfully protects against N-methyl aspartate neurotoxicity. Eur J Pharmacol. 1987;141:357–361. [CrossRef] [PubMed]
SiliprandiR, CanellaR, CarmignotoG, et al. N-methyl-D-aspartate-induced neurotoxicity in the adult rat retina. Vis Neurosci. 1992;8:567–573. [CrossRef] [PubMed]
HuangJC, SaltTE, VoadenMJ, MarshallJ. Non-competitive NMDA-receptor antagonists and anoxic degeneration of the ERG B-wave in vitro. Eye. 1991;5:476–580. [CrossRef] [PubMed]
CuiQ, HarveyAR. At least two mechanisms are involved in the death of retinal ganglion cells following target ablation in neonatal rats. J Neurosci. 1995;15:8143–8155. [PubMed]
OsborneNN, LarsenAK. Antigens associated with specific retinal cells are affected by ischaemia caused by raised intraocular pressure: effect of glutamate antagonists. Neurochem Int. 1996;29:263–270. [CrossRef] [PubMed]
FerreiraIL, DuarteCB, CarvalhoAP. Kainate-induced retina amacrine-like cell damage is mediated by AMPA receptors. Neuroreport. 1998;26:3471–3475.
OsborneNN, McCordRJ, WoodJPM. The effect of kainate on protein kinase C, GABA, and the uptake of serotonin in the rabbit retina in vivo. Neurochemistry. 1995;20:635–641. [CrossRef]
OsborneNN, LarsenA, BarnettNL. Influence of excitatory amino acids and ischemia on rat retinal choline acetyltransferase-containing cells. Invest Ophthalmol Vis Sci. 1995;36:1692–1700. [PubMed]
OsborneNN, HerreraAJ. The effect of experimental ischaemia and excitatory amino acid agonists on the GABA and serotonin immunoreactivities in the rabbit retina. Neuroscience. 1994;59:1071–1081. [CrossRef] [PubMed]
BrazeauP, ValeW, BurgusR, et al. Hypothalamic polypeptide that inhibits the secretion of immunoreactive pituitary growth hormone. Science. 1973;179:77–79. [CrossRef] [PubMed]
ShapiroB, KronheimS, PimstoneB. The presence of immunoreactive somatostatin in rat retina. Horm Metab Res. 1979;11:79–80. [CrossRef] [PubMed]
HoyerD, BellGI, BerelowitzM, et al. Classification and nomenclature of somatostatin receptors. Trends Pharmacol Sci. 1995;16:86–88. [CrossRef] [PubMed]
ThermosK. Functional mapping of somatostatin receptors in the retina: a review. Vision Res. 2003;43:1805–1815. [CrossRef] [PubMed]
HelboeL, MollerM. Immunohistochemical localization of somatostatin receptor subtypes sst1 and sst2 in the rat retina. Invest Ophthalmol Vis Sci. 1999;40:2376–2382. [PubMed]
JohnsonJ, WongH, WalshJH, BrechaNC. Expression of the somatostatin subtype 2A receptor in the rabbit retina. J Comp Neurol. 1998;393:93–101. [CrossRef] [PubMed]
JohnsonJ, WuV, WongH, WalshJH, BrechaNC. Somatostatin receptor subtype 2A expression in the rat retina. Neuroscience. 1999;94:675–683. [CrossRef] [PubMed]
VasilakiA, GardetteR, EpelbaumJ, ThermosK. NADPH-diaphorase colocalization with somatostatin receptor subtypes sst2A and sst2B in the retina. Invest Ophthalmol Vis Sci. 2001;42:1600–1609. [PubMed]
PetrucciC, RestaV, FieniF, BigianiA, BagnoliP. Modulation of potassium current and calcium influx by somatostatin in rod bipolar cells isolated from the rabbit retina via sst2 receptors. Naunyn Schmiedebergs Arch Pharmacol. 2001;363:680–694. [CrossRef] [PubMed]
VasilakiA, PapadakiT, NotasG, et al. Effect of somatostatin on nitric oxide production in human retinal pigment epithelium cell cultures. Invest Ophthalmol Vis Sci. 2004;45:1499–1506. [CrossRef] [PubMed]
CristianiR, FontanesiG, CasiniG, PetrucciC, ViolletC, BagnoliP. Expression of somatostatin subtype 1 receptor in the rabbit retina. Invest Ophthalmol Vis Sci. 2000;41:3191–3199. [PubMed]
VasilakiA, MouratidouM, SchulzS, ThermosK. Somatostatin mediates nitric oxide production by activating sst(2) receptors in the rat retina. Neuropharmacology. 2002;43:899–909. [CrossRef] [PubMed]
CristianiR, PetrucciC, Dal MonteM, BagnoliP. Somatostatin (SRIF) and SRIF receptors in the mouse retina. Brain Res. 2002;936:1–14. [CrossRef] [PubMed]
SmithLEH, KopchickJJ, ChenW, et al. Essential role of growth hormone in ischemia-induced retinal neovascularization. Science. 1997;276:1706–1709. [CrossRef] [PubMed]
HigginsRD, YanY, SchrierBK. Somatostatin analogs inhibit neonatal retinal neovascularisation. Exp Eye Res. 2002;74:553–559. [CrossRef] [PubMed]
ForloniG, LuccaE, AngerettiN, ChiessaR, VezzaniA. Neuroprotective effect of somatostatin on nonapoptotic NMDA-induced neuronal death: role of cyclic GMP. J Neurochem. 1997;68:319–327. [PubMed]
BraunH, SchulzS, BecherA, SchroderH, HolltV. Protective effects of cortistatin (CST-14) against kainate-induced neurotoxicity in rat brain. Brain Res. 1998;803:54–60. [CrossRef] [PubMed]
RaucaC, SchaferK, HolltV. Effects of somatostatin, octreotide and cortistatin on ischaemic neuronal damage following permanent middle cerebral artery occlusion in the rat. Naunyn Schmiedebergs Arch Pharmacol. 1999;360:633–638. [CrossRef] [PubMed]
ReinerPB, LaycockAG, DollCJ. A pharmacological model of ischemia in the hippocampal slice. Neurosci Lett. 1990;119:175–178. [CrossRef] [PubMed]
MastrodimouN, LambrouGN, ThermosK. Effect of somatostatin analogues on chemically induced ischaemia in the rat retina. Naunyn Schmiederbergs Arch Pharmacol. 2005;371:44–53. [CrossRef]
CatalaniE, CerviaD, MartiniD, et al. Changes in neuronal response to ischemia in retinas with genetic alterations of somatostatin receptor expression. Eur J Neurosci. 2007;25:1447–1459. [CrossRef] [PubMed]
MastrodimouN, ThermosK. The somatostatin receptor (sst1) modulates the release of somatostatin in rat retina. Neurosci Lett. 2004;356:13–16. [CrossRef] [PubMed]
SperkS, WidmannR. Somatostatin precursor in the rat striatum: changes after local injection of kainic acid. J Neurochem. 1985;45:1441–1447. [CrossRef] [PubMed]
BradfordMM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248–254. [CrossRef] [PubMed]
MasseySC, MillerRF. Glutamate receptors of ganglion cells in the rabbit retina: evidence for glutamate as a bipolar cell transmitter. J Physiol. 1988;405:635–655. [CrossRef] [PubMed]
YangXL. Characterization of receptors for glutamate and GABA in retinal neurons. Prog Neurobiol. 2004;73:127–150. [CrossRef] [PubMed]
HughesTE, Hermans-BorgmeyerI, HeinemannS. Differential expression of glutamate receptor genes (GluR1–5) in the rat retina. Vis Neurosci. 1992;8:49–55. [CrossRef] [PubMed]
PengYW, BlackstoneCD, HuganirRL, YauKW. Distribution of glutamate receptor subtypes in the vertebrate retina. Neuroscience. 1995;66:483–497. [CrossRef] [PubMed]
LuYM, YinHZ, ChiangJ, WeissJH. Ca(2+)-permeable AMPA/kainate and NMDA channels: high rate of Ca2+ influx underlies potent induction of injury. J Neurosci. 1996;16:5457–5465. [PubMed]
GeigerJR, MelcherT, KohDS, et al. Relative abundance of subunits mRNAs determines gating and Ca2+ permeability of AMPA receptors in principal neurons and interneurons in rat CNS. Neuron. 1995;15:193–204. [CrossRef] [PubMed]
JakobsTC, BenY, MaslandRH. Expression of mRNA for glutamate receptor subunits distinguishes the major classes of retinal neurons, but is less specific for individual cell types. Mol Vis. 2007;13:933–948. [PubMed]
FischerAJ, SeltnerRL, PoonJ, StellWK. Immunocytochemical characterization of quisqualic acid- and N-methyl-D-aspartate-induced excitotoxicity in the retina of chicks. J Comp Neurol. 1998;393:1–15. [CrossRef] [PubMed]
FirthSI, LiW, MasseySC, MarshakDW. AMPA receptors mediate acetylcholine release from starburst amacrine cells in the rabbit retina. J Comp Neurol. 2003;466:80–90. [CrossRef] [PubMed]
SagarSM. NADPH-diaphorase reactive neurons of the rabbit retina: differential sensitivity to excitotoxins and unusual morphologic features. J Comp Neurol. 1990;300:309–319. [CrossRef] [PubMed]
BluteTA, VelascoP, EldredWD. Functional localization of soluble guanylate cyclase in turtle retina: modulation of cGMP by nitric oxide donors. Vis Neurosci. 1998;15:485–498. [PubMed]
DijkF, KamphuisW. An immunocytochemical study on specific amacrine cell subpopulations in the rat retina after ischemia. Brain Res. 2004;1026:205–217. [CrossRef] [PubMed]
SinghM, SavitzSI, HoqueR, et al. Cell-specific caspase expression by different neuronal phenotypes in transient retinal ischemia. J Neurochem. 2001.466–475.
de la VillaP, KurahashiT, KanekoA. L-glutamate-induced responses and cGMP-activated channels in three subtypes of retinal bipolar cells dissociated from the cat. J Neurosci. 1995;15:3571–3582. [PubMed]
ShiellsRA, FalkG. The glutamate-receptor linked cGMP cascade of retinal on-bipolar cells is pertussis and cholera toxin-sensitive. Proc Biol Sci. 1992;247:17–20. [CrossRef] [PubMed]
KamphuisW, KloosterJ, DijkF. Expression of AMPA-type glutamate receptor subunit (GluR2) in ON-bipolar neurons in the rat retina. J Comp Neurol. 2003;455:172–186. [CrossRef] [PubMed]
LinB, MartinPR, GrunertU. Expression and distribution of ionotropic glutamate receptor subunits on parasol ganglion cells in the primate retina. Vis Neurosci. 2002;19:453–465. [PubMed]
BrandstätterJH, HartveitE, Sassoe-PognettoM, WässleH. Expression of NMDA and high-affinity kainate receptor subunit mRNAs in the adult rat retina. Eur J Neurosci. 1994;6:1100–1112. [CrossRef] [PubMed]
TezelG, WaxMB. Inhibition of caspase activity in retinal cell apoptosis induced by various stimuli in vitro. Invest Ophthalmol Vis Sci. 1999;40:2660–2667. [PubMed]
KataiN, YoshimuraN. Apoptotic retinal neuronal death by ischemia-reperfusion is executed by two distinct caspase family proteases. Invest Ophthalmol Vis Sci. 1999;40:2697–2705. [PubMed]
DijkF, Van LeeuwenS, KamphuisW. Differential effects of ischemia-reperfusion on amacrine-cell subtype specific transcript levels in rat retina. Brain Res. 2004;1026:194–204. [CrossRef] [PubMed]
CelikerU, IlhanN. Nitric oxide and octreotide in retinal ischemia-reperfusion injury. Doc Ophthalmol. 2002;105:327–338. [CrossRef] [PubMed]
TallentM, LiapakisG, O'CarrollA-M, LolaitSJ, DichterM, ReisineT. Somatostatin receptor subtypes SSTR2 and SSTR5 couple negatively to an L-type Ca2+ current in the pituitary cell line AtT-20. Neuroscience. 1996;71:1073–1081. [CrossRef] [PubMed]
MerineySD, GrayDB, PilarGR. Somatostatin-induced inhibition of neuronal Ca2+ current modulated by cGMP-dependent protein kinase. Nature. 1994;369:336–339. [CrossRef] [PubMed]
MastrodimouN, KiagiadakiF, ThermosK. The role of nitric oxide and cGMP in somatostatin’s protection against retinal ischemia. Invest Ophthalmol Vis Sci. 2008;49:342–349. [CrossRef] [PubMed]
OliasG, ViolletC, KusserowH, EpelbaumJ, MeyerhofW. Regulation and function of somatostatin receptors. J Neurochem. 2004;89:1057–1091. [CrossRef] [PubMed]
KeJB, ZhongYM. Expression of somatostatin receptor subtype 5 in rat retinal amacrine cells. Neuroscience. 2007;144:1025–1032. [CrossRef] [PubMed]
RohrerSP, BirzinET, MosleyRT, et al. Rapid identification of subtype-selective agonists of the somatostatin receptor through combinatorial chemistry. Science. 1998;282:737–740. [CrossRef] [PubMed]
BezerraY, FuselierJA, PeymanGA, OnerH, DrouantG, CoyDH. Study of inhibitory effects of an antioangiogenic somatostatin-camptothecin conjugate on laser-induced choroidal neovascularization in rats. Retina. 2005;25:345–354. [CrossRef] [PubMed]
HernandezC, CarrascoE, CasamitjanaR, DeulofeuR, García-ArumíJ, SimóR. Somatostatin molecular variants in the vitreous fluid: a comparative study between diabetic patients with proliferative diabetic retinopathy and nondiabetic control subjects. Diabetes Care. 2005;28:1941–1947. [CrossRef] [PubMed]
SimoR, CarrascoE, FonollosaA, García-ArumíJ, CasamitjanaR, HernándezC. Deficit of somatostatin in the vitreous fluid of patients with diabetic macular edema. Diabetes Care. 2007;30:725–727. [CrossRef] [PubMed]
PapadakiT, ZacharopoulosI, IaccheriB, FioreT, FosterCS. Somatostatin for uveitic cystoid macular edema (CME). Ocul Immunol Inflamm. 2005;13:469–470. [CrossRef] [PubMed]
PapadakiT, TsilimbarisM, ThermosK, et al. The role of lanreotide in the treatment of choroidal neovascularization secondary to age-related macular degeneration: a pilot clinical trial. Retina. 2003;23:800–807. [CrossRef] [PubMed]
RobertsonJE, WestraI, WolteringEA, et al. Intravitreal injection of octreotide acetate. J Ocul Pharmacol Ther. 1997;13:171–177. [CrossRef] [PubMed]
LiangC, PeymanGA, ConwayMD, WolteringEA. Retinal toxicity of intravitreous octreotide in the rabbit. Can J Ophthalmol. 1997;32:229–232. [PubMed]
Figure 1.
 
Dose-dependent effect of AMPA treatment on ChAT immunoreactivity in the retina. ChAT immunoreactivity in control rat retina (A) and in retina that received 21 (B), 42 (C), and 84 (D) nmol AMPA 1 day after injection. Partial loss of ChAT I-R was observed with 21 nmol/eye (only some processes of the cholinergic neurons appear), whereas the larger doses caused the complete loss of ChAT I-R. Scale bar, 50 μm.
Figure 1.
 
Dose-dependent effect of AMPA treatment on ChAT immunoreactivity in the retina. ChAT immunoreactivity in control rat retina (A) and in retina that received 21 (B), 42 (C), and 84 (D) nmol AMPA 1 day after injection. Partial loss of ChAT I-R was observed with 21 nmol/eye (only some processes of the cholinergic neurons appear), whereas the larger doses caused the complete loss of ChAT I-R. Scale bar, 50 μm.
Figure 2.
 
Time-dependent effect of AMPA treatment on ChAT immunoreactivity in the retina. ChAT immunoreactivity in rat retina that intravitreally received PBS (50 mM, control tissue; A) or AMPA (42 nmol/eye) 1 (B), 8 (C), 16 (D), and 30 (E) days after treatment. The complete loss of choline acetyltransferase expression from the first until the thirtieth day after treatment was observed. Arrows: nonspecific binding of the secondary antibody to blood vessels. Scale bar, 50 μm.
Figure 2.
 
Time-dependent effect of AMPA treatment on ChAT immunoreactivity in the retina. ChAT immunoreactivity in rat retina that intravitreally received PBS (50 mM, control tissue; A) or AMPA (42 nmol/eye) 1 (B), 8 (C), 16 (D), and 30 (E) days after treatment. The complete loss of choline acetyltransferase expression from the first until the thirtieth day after treatment was observed. Arrows: nonspecific binding of the secondary antibody to blood vessels. Scale bar, 50 μm.
Figure 3.
 
Effect of AMPA treatment on PKC and MAP expression in the retina. PKC immunoreactivity in control (A) and AMPA (42 nmol/eye)–treated tissue (B), MAP1A immunoreactivity in control (C), and AMPA-treated tissue 1 day after injection (D). Scale bar, 50 μm.
Figure 3.
 
Effect of AMPA treatment on PKC and MAP expression in the retina. PKC immunoreactivity in control (A) and AMPA (42 nmol/eye)–treated tissue (B), MAP1A immunoreactivity in control (C), and AMPA-treated tissue 1 day after injection (D). Scale bar, 50 μm.
Figure 4.
 
Effect of AMPA treatment on TH and recoverin expression in the retina. TH immunoreactivity in control (A) and AMPA (42 nmol/eye)–treated tissue and (B) recoverin immunoreactivity in control (C) and AMPA-treated tissue (D) 1 day after injection. Scale bar, 50 μm.
Figure 4.
 
Effect of AMPA treatment on TH and recoverin expression in the retina. TH immunoreactivity in control (A) and AMPA (42 nmol/eye)–treated tissue and (B) recoverin immunoreactivity in control (C) and AMPA-treated tissue (D) 1 day after injection. Scale bar, 50 μm.
Figure 5.
 
Effect of AMPA on bNOS immunoreactivity. bNOS-containing amacrine cells were counted in PBS and AMPA (42 nmol/eye)–treated retinal sections at 1, 8, 16, and 30 days. NOS-containing neurons were decreased 1 day after AMPA administration. However, the signal returned to control levels 8 days after AMPA injection. Results are presented as mean ± SEM.
Figure 5.
 
Effect of AMPA on bNOS immunoreactivity. bNOS-containing amacrine cells were counted in PBS and AMPA (42 nmol/eye)–treated retinal sections at 1, 8, 16, and 30 days. NOS-containing neurons were decreased 1 day after AMPA administration. However, the signal returned to control levels 8 days after AMPA injection. Results are presented as mean ± SEM.
Figure 6.
 
Effect of AMPA on SRIF levels. SRIF levels were measured by radioimmunoassay. No statistically significant changes were observed between the control and the AMPA (42 nmol/eye)–treated retinas 1 day after injection.
Figure 6.
 
Effect of AMPA on SRIF levels. SRIF levels were measured by radioimmunoassay. No statistically significant changes were observed between the control and the AMPA (42 nmol/eye)–treated retinas 1 day after injection.
Figure 7.
 
Effect of AMPA on sst2A expression levels. sst2A Immunoreactivity in PBS-treated and AMPA (42 nmol/eye)–treated retina 1 day after injection (A, B). sst2A Immunoreactivity is located in somata, axons, and processes of bipolar cells, and no differences between control and excitotoxic tissue were observed. Scale bar, 50 μm. Western blot studies on control retinal membranes (C). A protein of approximate molecular mass of 77 kDa was detected in the retina that was not visible in the presence of the sst2 antigen. No differences in the expression of the receptor protein (72 kDa) were observed in the PBS- and AMPA-treated retinas (D).
Figure 7.
 
Effect of AMPA on sst2A expression levels. sst2A Immunoreactivity in PBS-treated and AMPA (42 nmol/eye)–treated retina 1 day after injection (A, B). sst2A Immunoreactivity is located in somata, axons, and processes of bipolar cells, and no differences between control and excitotoxic tissue were observed. Scale bar, 50 μm. Western blot studies on control retinal membranes (C). A protein of approximate molecular mass of 77 kDa was detected in the retina that was not visible in the presence of the sst2 antigen. No differences in the expression of the receptor protein (72 kDa) were observed in the PBS- and AMPA-treated retinas (D).
Figure 8.
 
Effect of somatostatin on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with SRIF (10−5 M; C,10−4 M; D) 1 day after injection. Partial protection was afforded by SRIF 10−4 M. Scale bar, 50 μm.
Figure 8.
 
Effect of somatostatin on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with SRIF (10−5 M; C,10−4 M; D) 1 day after injection. Partial protection was afforded by SRIF 10−4 M. Scale bar, 50 μm.
Figure 9.
 
Effect of lanreotide (sst2/5 analog) on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with lanreotide (10−5 M; C, 10−4 M; D) 1 day after injection. Lanreotide protected the retina in a dose-dependent manner. Scale bar, 50 μm.
Figure 9.
 
Effect of lanreotide (sst2/5 analog) on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with lanreotide (10−5 M; C, 10−4 M; D) 1 day after injection. Lanreotide protected the retina in a dose-dependent manner. Scale bar, 50 μm.
Figure 10.
 
Effect of L-779976 (sst2 analog) on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with L-779976 (10−6 M; C, 10−5 M; D, 10−4 M; E) 1 day after injection. L-779976 protected the retina in a dose-dependent manner. Scale bar, 50 μm. (F) Effect of AMPA (42 nmol) alone or in combination with L-779976 (10−4 M) on the number of bNOS expressing retinal cells. The AMPA effect is statistically different when compared to the control (*P < 0.05).
Figure 10.
 
Effect of L-779976 (sst2 analog) on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with L-779976 (10−6 M; C, 10−5 M; D, 10−4 M; E) 1 day after injection. L-779976 protected the retina in a dose-dependent manner. Scale bar, 50 μm. (F) Effect of AMPA (42 nmol) alone or in combination with L-779976 (10−4 M) on the number of bNOS expressing retinal cells. The AMPA effect is statistically different when compared to the control (*P < 0.05).
Figure 11.
 
Effect of L-779976 (sst2 analog) on AMPA-induced apoptosis. TUNEL staining used to assess retinal cell death by AMPA and protection by the L-779976 analog 1 day after injection. AMPA afforded excitotoxic effects (B) compared with control (A). L-779976 (10−4 M) protected the retina from AMPA-induced apoptosis (C). Scale bar, 50 μm.
Figure 11.
 
Effect of L-779976 (sst2 analog) on AMPA-induced apoptosis. TUNEL staining used to assess retinal cell death by AMPA and protection by the L-779976 analog 1 day after injection. AMPA afforded excitotoxic effects (B) compared with control (A). L-779976 (10−4 M) protected the retina from AMPA-induced apoptosis (C). Scale bar, 50 μm.
Figure 12.
 
Effect of L-797591 (sst1 analog) and L-803087 (sst4 analog) ligands on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with L-797591 (10−4 M; C) or L-803087 (10−4 M; D) 1 day after injection. Activation of sst1 and sst4 receptors had no effect on the AMPA-induced loss of ChAT expression. Scale bar, 50 μm.
Figure 12.
 
Effect of L-797591 (sst1 analog) and L-803087 (sst4 analog) ligands on the AMPA-dependent loss of ChAT immunoreactivity in the retina. ChAT immunoreactivity in PBS-treated retina (A) and in retina that received AMPA (42 nmol) alone (B) or in combination with L-797591 (10−4 M; C) or L-803087 (10−4 M; D) 1 day after injection. Activation of sst1 and sst4 receptors had no effect on the AMPA-induced loss of ChAT expression. Scale bar, 50 μm.
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×