You are on page 1of 13

Experimental Eye Research 83 (2006) 758e770

www.elsevier.com/locate/yexer

A rat model of glaucoma induced by episcleral vein ligation


Saiyuu Yu, Teruyo Tanabe*, Nagahisa Yoshimura
Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Kyoto University, 54 Shogoin Kawahara-cho,
Sakyo-Ku, Kyoto 606-8507, Japan
Received 10 October 2005; accepted in revised form 21 March 2006
Available online 16 May 2006

Abstract

To establish a reliable animal model of glaucoma, we examined if episcleral vein ligation in rat eyes can induce intraocular pressure (IOP) el-
evation and concomitant characteristic morphological features of glaucoma. IOP elevation was detected on the next day (30.1  4.4 mmHg:
operated eyes; 21.0  1.8 mmHg: control eyes) and persisted at least 7 months after the procedure (24.5  2.3 mmHg: operated eyes;
19.7  1.9 mmHg: control eyes). These results suggest that episcleral vein ligation can induce very mild IOP elevation immediately after the
operation, which can last over several months. Furthermore, it appears there was little variability in the patterns of IOP elevation among the
individual eyes treated with episcleral vein ligation. Morphological changes were detected selectively in the retinal ganglion cell (RGC) layer
and optic disc excavation was evident in the late stage of chronic IOP elevation. RGCs were selectively lost by apoptotic death. The number of
RGCs was reduced by 18% at 12 weeks and eventually by 35% at 8 months postoperatively. Müller cells downregulated the expression of
p27Kip1 and appeared to be partially in a reactive state even at the advanced stages of glaucoma. The expression of basic fibroblast growth
factor and ciliary neurotrophic factor, which are neurotrophic factors implicated in the control of cell survivals and neuroprotection, significantly
declined at the advanced stages. Taken altogether, these observations indicate that the episcleral vein ligation model based on the simple ligation
procedure reproducibly provides a reliable glaucoma model and contributes to give insights into the underlying molecular and cellular bases of
human glaucoma and to devise the new medication upon the disease.
Ó 2006 Elsevier Ltd. All rights reserved.

Keywords: glaucoma; rat model; episcleral vein ligation; retinal ganglion cell; apoptosis; trophic factor; Müller cell

1. Introduction of retina and optic disc are the most similar to human; however,
the costs are higher in preparing the samples compared to other
Glaucoma is an optic neuropathy characterized by a slow pro- species (Pederson and Gaasterland, 1984; Glovinsky et al.,
gressive loss of retinal ganglion cells (RGCs) and excavation of 1991; Quigley et al., 1995). In contrast, models using rodents
the optic disc (Quigley and Green, 1979; Quigley et al., 1988; are advantageous in that large numbers of samples can be pre-
Glovinsky et al., 1991). Elevation of intraocular pressure pared and handled at a time. In addition, spontaneous or genet-
(IOP) has been implicated as the most critical risk factor in ically manipulated mutant mice, which exhibit elevation of IOP,
the generation of glaucomatous optic neuropathy. Several ani- are available (John et al., 1998; Grozdanic et al., 2003b;
mal models have been devised to elucidate the molecular and Mabuchi et al., 2004). However, these mice only manifest the
cellular bases of how IOP elevation triggers RGC death and glaucoma phenotype after a long period of time and current
how RGC death progresses to glaucoma, and also to explore IOP measurement techniques in mice are invasive. On the other
treatments to inhibit the death of RGCs. A model of glaucoma hand, in rats, operational procedures are applicable and IOP
using monkey is beneficial in that the gross anatomical bases measurement are non-invasive; thus, a wide variety of rat glau-
coma models, using hypertonic saline injection, cauterizing
episcleral veins or laser photocoagulation, have been devised
* Corresponding author. Fax: þ81 75 752 0933. and analyzed (Shareef et al., 1995; Morrison et al., 1997;
E-mail address: tanabet@kuhp.kyoto-u.ac.jp (T. Tanabe). Ueda et al., 1998; Levkovitch-Verbin et al., 2002). Every model

0014-4835/$ - see front matter Ó 2006 Elsevier Ltd. All rights reserved.
doi:10.1016/j.exer.2006.03.014
S. Yu et al. / Experimental Eye Research 83 (2006) 758e770 759

shows a significant IOP elevation, glaucomatous changes in the measured in the operated eyes was found to be less than
retina and is valuable as a glaucoma model; however, some 25 mmHg at 2 weeks or at 4 weeks after the initial ligation,
models require special devices or techniques to induce repro- we further ligated newly formed collateral veins through which
ducible IOP elevation. blood flow escapes and thus subdivided all the treated samples
To establish a reliable rat model that exhibits reproducible into three groups [Group 1, single treatment (n ¼ 29); Group 2,
and consistent IOP elevation over a long period of time with retreatment at 2 weeks (n ¼ 18); Group 3, retreatment at
simple procedure, we performed episcleral vein ligation and ob- 4 weeks (n ¼ 24)] to analyze the level of IOP.
served whether IOP was elevated consistently over a long pe-
riod, and if any morphological changes, including RGC loss, 2.3. Intraocular pressure monitoring
resembling glaucomatous phenotypes in human, were induced.
IOP was measured using a digital tonometer (Tonopen XL,
Mentor; Norwell, MA, USA) under general and local anesthe-
2. Materials and methods sia as described above. General anesthesia is known to affect
the level of IOP (Krupin et al., 1980; Jia et al., 2000). There-
2.1. Animals fore, to circumvent the potential effect of general anesthesia,
we conducted IOP measurements as soon as rats were lightly
All the animal experiments were performed in compliance sedated by anesthesia. The automatic average values of accept-
with the ARVO Statement for the Use of Animals in Ophthalmic able several consecutive measurements, with a coefficient of
and Vision Research. Adult female Wister rats, 9 weeks of age, variation less than 5%, was recorded three times and averaged.
were used in this study. Rats were maintained in an environmen- Furthermore, we also evaluated the Tonopen XL readings in
tally controlled room with 14-h light/10-h dark daily cycles. relation to the transducer pressure according to the method
previously described (Mermoud et al., 1994). A regression
2.2. Rat model of chronic ocular line calculated was y ¼ 2.8533 þ 0.8232x (r2 ¼ 0.959). The
hypertension (glaucoma) values used in this study are actual readings by Tonopen and
no correction with a pressure transducer was performed. We
Rats were lightly anesthetized by intraperitoneal injection of measured IOP of both eyes on the next day, day 3, weekly
a mixture of ketamine (25 mg/kg) and xylazine (10 mg/kg); the for the first 3 months and monthly afterwards. All measure-
eyes were further anaesthetized locally with a topical applica- ments were started at 10:00 h during the follow-up period
tion of 0.5% proparacaine (Santen) eyedrop. Small incisions and were basically conducted by one examiner. Another exam-
into the conjunctiva and Tenon’s capsule covering the episcleral iner selected samples randomly and measured IOP in a blind
veins in each quadrant of the right eye were made and the vein fashion. The difference in IOP values between two exam-
was exposed. Ligation was applied to three trunks of the epis- iners/the average IOP of two examiners were 3.5  2.1%.
cleral veins and a branch of the other episcleral veins in the right
eye (Fig. 1) using 10-0 nylon (Alcon surgical). On the left eye, 2.4. RGC labeling and counting
sham surgery was performed in the same manner except for li-
gating episcleral veins. Antibiotic ointment was applied topi- Retrograde labeling and counting of RGCs was performed
cally after each procedure. When the mean value of IOP according to the procedure described previously (Vidal-Sanz

Fig. 1. Anatomical bases of the operation of episcleral vein ligation in rat eyes. Topographical arrangement of episcleral/orbital vein (thick red line), limbal vein
(thin red line surrounding cornea), cornea, and rectus muscles (double thin black lines) is indicated in scheme on the left. The site of ligation is indicated by double
thick black lines on the episcleral vein. Picture on the right shows an example of operated eyes in which an episcleral/orbital vein was ligated. The central side of
the ligated vein became congested. This congestion is a criterion showing if the vein is ligated sufficiently to obstruct the blood flow. SR, superior rectus muscle;
SO, superior oblique muscle; MR, medial rectus muscle; LR, lateral rectus muscle; IR, inferior rectus muscle; IO, inferior oblique muscle.
760 S. Yu et al. / Experimental Eye Research 83 (2006) 758e770

et al., 1988). At various time points after episcleral vein liga- CD11B (OX42) (1:1000; Chemicon international, Temecula,
tion (6, 12, 18, 24 and 32 weeks; n ¼ 5 per each time point), CA, USA), human polyclonal antibody against p75NGF
the rats under deep anesthesia (mixture of ketamine (1:1000; Promega, Madison, WI, USA; Cappel, Durham, NC,
(37.5 mg/kg) and xylazine (15 mg/kg)), were immobilized ste- USA), mouse monoclonal antibody against protein kinase C
reotaxically and were microinjected with 2.0 ml of a 5% solu- (PKC) (1:100; Sigma, St. Louis, MO, USA), mouse monoclonal
tion of fluorescent tracer 1,10 -dioctadecyl-3,3,30 ,30 -tetramethyl antibody against opsin (1:8000; Sigma), rabbit polyclonal anti-
indocarbocyanine perchlorate ((DiI; Molecular Probes, Eu- body against calbindin D-28K (1:200; Chemicon International),
gene, OR, USA) in distilled water bilaterally into the superior mouse monoclonal antibody against glial fibrillary acidic pro-
colliculus (SC). Seven days after DiI injections, eyes were har- tein (GFAP) (1:1000; Sigma), mouse antibody against
vested after perfusion with 4% PFA, and the isolated retinas p27Kip1 (1:200; BD Transduction Laboratories, San Jose,
were flat-mounted, with vitreous side up, on a glass slide, CA, USA), goat polyclonal antibody against Brn-3b (C-13)
air-dried and mounted in Fluorosave (Calbiochem, La Jolla, (1:100; Santa Cruz Biotechnology, Santa Cruz, CA, USA),
CA, USA). Peripapillary (PP: 1 mm from the optic disc), mid- sheep polyclonal antibody against fibroblast growth factor, basic
dle (M: 2 mm from the optic disc) and peripheral (PR: 3 mm (bFGF) (1:1000; Chemicon), sheep polyclonal antibody against
from the optic disc) retinal areas per each retinal quadrant, 12 brain derived neurotrophic factor (BDNF) (1:1000; Chemicon),
areas in total, were subjected to RGC counting. Each rectangu- sheep polyclonal antibody against glial derived neurotrophic
lar area measured was 0.3257  0.3257 mm. The DiI-labeled factor (GDNF) (1:1000; Chemicon), mouse monoclonal anti-
RGCs were analyzed by a confocal microscope (CLSM; body against ciliary neurotrophic factor (CNTF) (1:100; Chem-
Carl Zeiss) and counted with a computer-assisted image- icon). For secondary antibodies, Alexa Fluor 488-conjugated
processing unit and the ‘‘count-measure’’ operating software anti-mouse, anti-rabbit, anti-sheep and anti-goat IgG (Molecu-
(Image-Pro Plus software; Media Cybernetics, Silver Spring, lar Probes) were used. TO-PRO-3 (Molecular Probes) was
MD, USA). To exclude the microglia, which are fluores- used for the nucleic acid stains. Immunofluorescence was ana-
cence-labeled by phagocytosing dying labeled RGCs, DiI- lyzed by confocal microscopy (Carl Zeiss).
labeled cells smaller than 10 mm2 or with a spindle-shape
were not counted (Naskar et al., 2002). Data represents the 2.7. Real-time RTePCR
number of RGCs per square millimeter.
Ten, 20 and 30 weeks postoperatively, rats were anesthetized
2.5. TUNEL (TdT-dUTP terminal nick-end with ketamine/xylazine and perfused intracardially with medi-
labeling) assay cal grade physiological saline (n ¼ 5 per each time point). The
eyes were enucleated, anterior segments were removed, and
After 6, 12 and 18 weeks of the initial episcleral vein liga- whole retinas were isolated. Total RNA was extracted from ret-
tion, a TUNEL assay was performed on the treated and control inas with the Qiagen RNeasy mini columns (Qiagen Inc.,
retinas that were retrogradely labeled by DiI 1 week before the Valencia, CA) according to the manufacture’s protocol. The
assay (n ¼ 5 per each time point). Eyes were fixed by perfu- concentration and purity of the extracted RNA were evaluated
sion and DNA nick-end labeling was performed on flat- by optical density measurements at 260 nm and 280 nm. Ran-
mounted retina using a Mebstain apoptosis kit (MBL, Nagoya, dom-primed cDNA was synthesized from 1 mg total RNA using
Japan), according to the manufacturer’s protocol. TUNEL- the First-Strand cDNA Synthesis kit (Amersham Pharmacia
positive DiI-labeled cells were analyzed by a confocal micro- Biotech, Uppsala, Sweden). Quantitative PCR was carried out
scope (CLSM; Carl Zeiss) and the number of apoptotic cells in in 96-well optical reaction plates using real-time TaqManÒ
the entire retina was analyzed quantitatively with a computer- technology and the results were analyzed with a Model 7000
assisted image-processing unit and ‘‘count-measure’’ operat- Sequence Detector System (Applied Biosystems, Foster City,
ing software (Image-Pro Plus software; Media Cybernetics). CA). The mixed real-time PCR solution, 25 ml in total, con-
tained 2 TaqMan Universal PCR Mastermix (Applied Biosys-
2.6. Histological and immunohistochemical analyses tems), 1 mM of each primer, 250 nM of TaqMan probes, and
25 ng of each cDNA. The thermal cycling conditions comprised
Ten, 20 and 30 weeks postoperatively, the treated and control 50  C for 2 min, 95  C for 10 min, and 45 cycles of 95  C for
eyes (n ¼ 3 per each time point, respectively) were paraffin- 15 s plus 60  C for 1 min, using the standard protocol of the
embedded and histological changes were analyzed by light manufacturer. Quantitative normalization in each sample was
microscopy, as described previously (Miyawaki et al., 2004). performed using the expression of rodent glyceraldehyde-
Immunohistochemical analyses for retinal cell-specific markers 3-phosphate dehydrogenase (GAPDH) gene as an internal con-
were performed on control and glaucoma retinas 32 weeks after trol. The primers and probes for bFGF were designed with
the treatment (n ¼ 3, respectively), while analyses for neurotro- primer-express software (Applied Biosystems) as follows:
phic factors were performed on 10-, 20- and 30-week glaucoma bFGF. Forward, 50 -GAACGCCTGGAGTCCAATAACTA-30 ,
and control retinas (n ¼ 4 per each time point, respectively). Im- reverse, 50 -CCCGTTTTGGGATCCGAGTTT-30 , detection probe,
munohistochemistry was performed on cryosections according 50 -ACACTTACCGGTCACGGAAATACTCCAGTT-30 . The primers
to the method described previously (Miyawaki et al., 2004). and probes used for GFAP, p27kip1, GDNF, CNTF, BDNF
The antibodies used were: mouse monoclonal antibody against and GAPDH were from Applied Biosystems.
S. Yu et al. / Experimental Eye Research 83 (2006) 758e770 761

2.8. Statistical analysis 3.2. Morphological changes of the retinas with


elevated IOP
Data are expressed as mean  S.D. (IOP recordings) and
mean  S.E.M. (number of RGC and real-time RTePCR). We next examined if there were any morphological changes
Data between groups were compared with Student’s t-test or in retinas in relation to the elevated IOP induced by episcleral
repeated measures ANOVA with Dunnett’s multiple compari- vein ligation (Fig. 3). The posterior eye segments (treated
son tests. Statistical significance was declared for P < 0.05. eyes: n ¼ 6; control eyes: n ¼ 6), including retinas and optic
discs, were histologically analyzed by light microscopy at early
3. Results (10 weeks), middle (20 weeks) and late (30 weeks) stages of
chronic IOP elevation. In samples analyzed 10 weeks after
3.1. Intraocular pressure was consistently elevated by treatment, both the retina and optic disc of treated eyes showed
episcleral vein ligation no apparent morphological difference from the control eyes
(Fig. 3AeD). At 20 weeks postoperatively, the reduction in
We first examined the temporal changes of IOP over several the number of RGCs was detected histologically, while the mor-
months in the operated and sham-operated groups, and analyzed phology of optic disc was similar to the control (Fig. 3EeH). At
the level, persistency and reproducibility of IOP elevation 30 weeks after treatment, a marked decrease in RGC numbers
(Fig. 2). All the values presented in this study are the actual read- was observed (Fig. 3I,J). In addition, a significant excavation
ings measured by Tonopen XL. The baseline IOP before treat- was observed in the optic disc of the treated eye (Fig. 3K,L).
ment was 19.9  1.9 (mean  S.D.) mmHg. On the next day Apart from the morphological changes observed in the ganglion
after the surgery, all treated eyes showed elevated IOP cell layer (GCL), the outer and inner nuclear layers (ONL and
(30.1  4.4 mmHg) compared to controls (21.0  2.0 mmHg). INL) did not exhibit any difference between the control and
On the third day after the surgery, all treated eyes showed treated eyes throughout the follow-up period (Fig. 3).
elevated IOP (27.7  3.6 mmHg) compared to controls
(20.9  1.9 mmHg). At 1 week after1 the surgery, all treated 3.3. Spatial and temporal patterns of reduction
eyes showed elevated IOP (27.8  1.3 mmHg) compared to con- in the number of RGCs
trols (20.2  1.5 mmHg). In 40.8% of glaucoma model speci-
mens we examined, the initially elevated IOP persisted for as To monitor the extent and the time course of the loss of RGC
long as 7 months (Table 1). On the other hand, in 59.2% of spec- in a more quantitative manner, RGCs were selectively labeled
imens, initially elevated IOP showed a significant decline by retrograde dye tracing from the SC using DiI and the number
(25 mmHg) within 2e4 weeks, probably because of the newly of RGCs was analyzed by DiI fluorescence. We performed
formed collateral vessels. Further ligation was performed in quantitative analyses of the number of DiI-labeled RGCs that
those specimens at the second or fourth week after the first liga- are located in each quadrant of retinas of treated and control
tion. These subsequent ligation procedures induced persistent eyes derived at 6, 12, 18, 24 and 32 weeks after the treatment
IOP elevation afterwards in all samples (Table 1). The level, per- (n ¼ 5 per each case) (Fig. 4, Table 2). TUNEL assay was
sistency and temporal changes of IOP elevation observed in the also performed on whole-mount retinas to see if the loss of
‘‘double-treated’’ group were essentially similar to those ob- RGC is mediated by apoptosis (Fig. 5). When the number of
served in the ‘‘single-treated’’ group during the chronic phase RGCs at 6 weeks was analyzed, no significant change in the
of IOP elevation. At 7 months, the IOP measured was number was observed between the treated and control retinas
24.7  2.2 mmHg in the operated eyes (single or double treated) (P ¼ 0.68). At 12 weeks, the number of RGCs in the treated ret-
and 19.7  1.9 mmHg in the sham-operated eyes (P < 0.001). inas gradually decreased, by 18% in total. It appeared there was
no regional difference for the RGC loss between quadrants of
the retina (Fig. 5A). In contrast, the extent of RGC loss is pref-
erentially high in the peripapillary retina (20.52  1.70% re-
duction) compared to the peripheral retina (13.12  2.83%
reduction) (Fig. 4, Table 2). At 24 weeks, nearly 35% reduction
in the RGC number was detected in the treated retinas com-
pared to the controls, and it appeared that there was no further
reduction in the number of RGC from 24 weeks onward. Our
results show that the number of TUNEL-positive DiI-labeled
RGCs detected in the treated retinas was significantly high
(32.8  9.5, 26.4  4.7 and 30.8  3.8 cells per entire retina
Fig. 2. Temporal patterns of IOP elevation after the initial episcleral vein liga- at 6, 12 and 18 weeks, respectively: P ¼ 0.001) compared to
tion. Averages of IOP levels in the operated eyes (single and double treated) 1, the controls (8.4  1.0, 6.8  1.4 and 2.4  0.5 cells per entire
3, 6, 12, 24 and 32 weeks after treatment were 27.8, 26.6, 27.1, 27.1, 26.5 and retina at 6, 12 and 18 weeks, respectively) (Fig. 5B).
22.4 mmHg, respectively. IOP of the glaucoma eyes (black line) are signifi-
cantly higher than those of the sham-operated control eyes (dotted line) until We further analyzed quantitatively if there is any size-
7 months after operation. By the end of 32 weeks, IOP of operated eyes de- dependent preferential loss of RGC in the glaucomatous ret-
clines to control levels. Data expressed are mean  S.D. (mmHg). inas induced by episcleral vein ligation at 12 and 24 weeks
762 S. Yu et al. / Experimental Eye Research 83 (2006) 758e770

Table 1
Postoperative IOP of episcleral vein ligation model (mmHg  S.D.)
Treatment type Postoperative IOP (mmHg  SD)
1 week 2 weeks 4 weeks 12 weeks 28 weeks
Group 1 G 27.8  1.2 29.1  1.8 27.5  1.4 27.4  1.2 24.3  2.3
(single treatment: n ¼ 29) C 20.1  1.3 20.0  1.5 19.9  1.1 19.9  1.3 19.8  1.5
Group 2 G 28.1  1.3 24.3  0.6 28.1  0.8 26.9  1.7 26.2  1.5
(retreatment at 2 weeks: n ¼ 18) C 20.5  1.7 20.4  1.5 20.1  1.5 20.3  0.8 20.1  1.5
Group 3 G 27.6  1.4 28.3  1.4 24.6  0.6 26.9  1.4 24.6  2.1
(retreatment at 4 weeks: n ¼ 24) C 19.9  1.5 20.2  1.7 20.2  1.7 20.1  1.0 19.8  1.3
G, glaucoma model; C, control.

postoperatively (Fig. 6). The cell loss was preferentially ob- The intensity of expressions of p75 and Brn3b, specific molec-
served in RGC population with the diameter range between ular markers for RGCs, was decreased at 32 weeks, consistent
5 and 15 mm in the treated retinas compared to the controls with the reduction in the number of retrogradely labeled
at 12 and 24 weeks. In contrast, RGCs with the diameter larger RGCs. In contrast, PKC (a bipolar cell marker), opsin (a rod
than 15 mm did not show any decrease in their numbers by the photoreceptor marker) and calbindin (an amacrine and a hori-
treatment. A similar tendency of size-dependent RGC loss was zontal cell marker) were detected in a similar pattern in their
observed at early (12 weeks) and late (24 weeks) stages of IOP expression in the treated retina compared to the control.
elevation (Fig. 6). At 24 weeks, RGCs with diameter less than Among the glial cell markers, the expression of GFAP, an as-
5 mm showed statistically significant decrease in the treated trocyte marker that is expressed in reactive Müller glia under
retinas compared to the controls. pathological circumstances, appeared to be increased com-
pared with control retina. In addition, the expression of
3.4. Selective impairment of RGCs and Müller cells p27Kip1, a cyclin-dependent kinase inhibitor that is expressed
revealed by immunohistochemical analyses and by normal Müller glia but is downregulated under the reactive
real-time RTePCR condition, appeared to be decreased in the treated retina. To
further analyze the level of GFAP and p27Kip1 in a quantita-
To further analyze if there was any other cellular defects in tive manner, we performed a series of real-time PCR experi-
the retina at later stages of IOP elevation, immunohistochem- ment to analyze the level of each mRNA expressed in
ical analysis was performed using antibodies that are specific treated and control retinas after 32 weeks of surgery. Our re-
for each retinal cell type after 32 weeks of surgery (Fig. 7). sults showed that there was no difference in the expression

Fig. 3. Light microscopic analysis of control and glaucoma model eyes at 10 (AeD), 20 (EeH) and 30 (IeL) weeks after treatment. See details in the text. During
the 10e30-week period, a gradual loss of RGCs (B, F, J) and an augmentation of excavation of optic disc (D, H, L) in the operated eyes are detected. Sham-
operated control eyes showed no apparent morphological changes (A, C, E, G, I and K). Optic disc excavation (*) is evident in 30w glaucoma eye (L). GCL,
retinal ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer; Gla, glaucoma model. Scale bar: 50 mm.
S. Yu et al. / Experimental Eye Research 83 (2006) 758e770 763

Fig. 4. Temporal patterns of RGC loss in each region of the retina in operated and control eyes after treatment. RGCs (per square millimeter) were counted by ret-
rogradely transferred fluorescent dye. (A) Changes in the number of RGCs in the peripapillary (PP) region surrounding the optic disc at 1 mm radius. (B) Changes in
the number of RGCs in the midperipheral (M) region surrounding the optic disc at 1 mm width located 1e2 mm apart from the center of the optic disc. (C) Changes in
the number of RGCs in the peripheral (PR) region surrounding the optic disc at 1 mm width located 2e3 mm apart from the center of the optic disc. (D) Changes of the
total number of RGCs from the peripapillary through the peripheral region of retinas. From 12 weeks onward, a significant reduction in the number of RGCs is de-
tected in every region. Nearly 35% of RGCs were lost in total by 32 weeks. Data expressed are mean  S.E.M. (cell number/mm2).

level of GFAP in the treated and non-treated retinas, whereas trophic factors exhibited changes in their expressions during
the level of p27Kip1 was significantly reduced in the treated the period of IOP elevation. CNTF expression, preferentially
retinas (Fig. 8). detected in the GCL in the control retinas, is detected also
in the INL at 10 weeks but declined at 30 weeks in the glau-
3.5. Changes in the expression pattern of coma retina (Fig. 9AeD). Consistent with the decline in the
neurotrophic factor CNTF expression at 30 weeks, the quantitative analyses of
CNTF mRNA using real-time PCR showed a significant de-
To monitor if there were any changes in the expression of crease at 30 weeks after IOP elevation (Fig. 10). Basic FGF
neurotrophic factors, we next analyzed the expression of neu- expression, detected in the NFL, GCL and INL in the control
rotrophic factors, BDNF, CNTF, GDNF and basic FGF, which retina, showed no change at 10 weeks (Fig. 9F), declined in
are implicated in the control of RGC survival in previous stud- the GCL but maintained in the INL and NFL at 20 weeks
ies (Johnson et al., 1986; Mey and Thanos, 1993; Sapieha (Fig. 9G), and was markedly reduced in all the layers at
et al., 2003; Ji et al., 2004; Wu et al., 2004) (Fig. 9). We fo- 30 weeks (Fig. 9H). Consistent with this observation, total
cused our analyses on retinas that were derived from early amount of basic FGF mRNAs decreased significantly at
(10 weeks), middle (20 weeks) and late (30 weeks) stages of 30 weeks postoperatively (Fig. 10). BDNF and GDNF, both
chronic IOP elevation. When control retinas were immunohis- of which are mainly expressed in the GCL, did not change
tochemically analyzed, there was no change in the expression their expressions throughout the entire period of IOP elevation
patterns of trophic factors throughout the follow-up period (Fig. 9IeP), which is consistent with the quantitative analyses
(Fig. 9A,E,I,M). In contrast, in glaucoma retina, some of the of BDNF and GDNF mRNA expressions (Fig. 10).

Table 2
% Reduction of RGC in glaucoma model compared to control retinas
Area % Reduction of RGC
6 weeks 12 weeks 18 weeks 24 weeks 32 weeks
PP 3.62  3.02 20.52  1.70 26.17  2.55 33.89  2.74 33.26  2.39
M 0.98  2.72 19.00  1.65 26.10  2.02* 32.84  2.37 36.01  2.11
PR 4.73  4.47 13.12  2.83** 16.64  4.70 29.76  3.40 30.07  3.29
Total 3.12  1.95 17.55  1.26 22.97  1.93 32.16  1.61 33.11  1.51
Data are the mean  S.E.M. *P < 0.05, between M and PR; **P < 0.05, between PP and PR.
764 S. Yu et al. / Experimental Eye Research 83 (2006) 758e770

Fig. 5. Apoptosis in glaucoma model retina detected by terminal dUTP nick-end labeling (TUNEL). (A) Confocal microscopic analyses of RGC death detected by
a combination of TUNEL staining (green) and retrograde fluorescent dye tracing (red). Each picture represents a region of whole-mount retina of 18 weeks after the
treatment. N, nasal; S, superior; T, temporal; I, inferior; PR, peripheral; PP, peripapillary. TUNEL-positive cells are confined to the retrogradely labeled RGCs in
the glaucoma retina (bar ¼ 50 mm). (B) Table showing the number of apoptotic cells detected in the control and glaucoma model retina at 6, 12, and 18 weeks after
treatment. The number of TUNEL-positive cells in glaucoma rat retinas is more than 10 times than that in sham-operated control retinas (n ¼ 5).

4. Discussion the cause of IOP elevation. Instead of cautery burn, we utilized


a 10-0 nylon suture to obstruct the episcleral vein. One of the
4.1. Features of the episcleral vein ligation most distinguished features of our model is the very mild ele-
glaucoma rat model vation of IOP that can be induced immediately after the oper-
ation and can be persisted over several months after the single
Previous in vivo studies of glaucoma utilized many ap- or double treatments. Furthermore, there was little variability
proaches to induce IOP elevation in rat eyes. These studies in the patterns of IOP elevation among the eyes treated with
showed that most procedures were able to induce a significantly episcleral vein ligation. Our glaucoma model was capable of
high IOP, a more than 1.5 times increase compared to the con- inducing mild IOP elevation on the next day immediately after
trol eyes (Shareef et al., 1995; Morrison et al., 1997; Ueda the treatment and maintaining its elevation for as long as
et al., 1998; Levkovitch-Verbin et al., 2002), which mostly 7 months: w25 mmHg in the treated eyes and w20 mmHg
lasted for 1e3 months (Morrison et al., 1997; Levkovitch- in the control eyes. The levels of IOP at chronic phase were
Verbin et al., 2002) and are correlated with morphological maintained at a similar extent among the operated samples.
changes similar to those observed in human glaucoma subjects. Nearly 40% of the operated eyes showed elevated IOP consis-
In addition, these glaucoma models provided further insights tently only by a single ligation procedure. The rest (w59.2%)
into the cellular defects induced by chronic IOP elevation of the operated eyes, in contrast, did not exhibit IOP elevation
and beneficial for our understanding of the process of glau- continuously after initial treatment, however, a subsequent re-
coma. Among the glaucoma models, Shareef’s model is one ligation procedure was sufficient for the achievement and
of the most established glaucoma rat models, which obstructs maintenance of the IOP elevation in all the cases. Other
two or three trunks of episcleral veins by cauterization to models also reported the requirement of multiple retreatment
induce high IOP. Several laboratories have conducted studies to maintain IOP elevation (Morrison et al., 1997; Ueda
on glaucoma using this model; however, the IOP profiles et al., 1998; Mittag et al., 2000): 94.7% of our model showed
in those studies varied between laboratories (Sawada and consistent IOP elevation for at least 7 months with one addi-
Neufeld, 1999; Mittag et al., 2000; Grozdanic et al., 2003a; tional treatment. Indeed, the 10-0 nylon suture, which was
Kanamori et al., 2004) and it was technically difficult to pro- visible through the conjunctiva, enabled us to distinguish col-
duce this model in our experience. These observations promp- lateral veins that needed to be ligated at the time of re-
ted us to establish another glaucoma model. treatment from the ones that have been ligated previously.
Our glaucoma model is essentially similar to the episcleral Compared to laser photocoagulation or hypertonic saline
vein cautery model in that the obstruction of episcleral vein is injection models (Morrison et al., 1997; Levkovitch-Verbin
S. Yu et al. / Experimental Eye Research 83 (2006) 758e770 765

A previous study showed that there was a preferential cell


loss in the peripheral regions of retinas (Garcia-Valenzuela
et al., 1995; Laquis et al., 1998; Sawada and Neufeld, 1999).
Other studies showed a preferential RGC loss in the temporal
upper and lower regions of retinas (Morrison et al., 1997; Mit-
tag et al., 2000). In contrast, our ligation model exhibited the
susceptibility for the cell loss in the parapapillary retina at
early stage after IOP elevation. These apparent distinct fea-
tures of preferential regional sensitivity for cell loss in our
model might be due to the mild extent and consistency of
IOP elevation observed in our model. In addition to the re-
gional preference regarding RGC loss in glaucoma, the sus-
ceptibility of RGCs to IOP elevation depending on the size
is also the important issue. Several groups have shown that
larger ganglion cells are more vulnerable to IOP elevation in
a variety kind of species (Glovinsky et al., 1991; Danias
et al., 2006; Levkovitch-Verbin et al., 2002). However, the
preferential loss of RGCs in large size was nor detectable in
our episcleral vein ligation model. Further studies are needed
to see how regional distinct susceptibility is induced by the
different approaches to elevate IOP and the different pattern
of IOP elevation.
Previous studies using episcleral vein cautery model sug-
gested that the occlusion of episcleral vein might lead to the
impairment of ocular blood flow and the choroidal insuffi-
ciency, which may cause secondary effect on retina, especially
Fig. 6. Size distribution of DiI labeled RGCs in control and treated (Gla on photoreceptors. Grozdanic et al. reported the change in
model: 12w, 24w) retinas as a function of the cell diameter. RGCs with the ERG parameters; however, these changes were observed
diameter ranging between 5 and 15 mm appear to be preferentially decreased only during the period of IOP elevation and there was no
in the treated retinas compared to the controls. In contrast, RGCs larger than change in the cytoarchitectures of treated retinas (Grozdanic
15 mm in diameter did not show significant decrease in their numbers by the
et al., 2003a). Bayer et al. also reported the changes in the sco-
treatment. This tendency of size-dependency of RGC loss was observed
both at 12 and 24 weeks postoperatively. At 24 weeks, RGCs with diameter topic ERG in their modified episcleral vein ligation model, but
less than 5 mm showed statistically significant decrease in the treated retinas suggest that changes in the patterns of ERG appear to be
compared to the controls. mostly mediated by IOP elevation but not by the impairment
of ocular blood flow (Bayer et al., 2001). Other studies on
et al., 2002), the suturing approach utilized in our model did the episcleral vein cautery model also report that only RGCs
not induce any visible inflammation in the eyes even with re- are affected. In our present study, by ligating episcleral vein,
treatment (data not shown). we cannot rule out the possibility of impairment of ocular
blood flow and choroidal insufficiency. However, we observed
that there was a pronounced blood flow through the unligated
4.2. Episcleral vein ligation model recapitulates veins after the treatment, which might reflect the existence of
characteristic morphological features of glaucoma compensational shunt of blood flow via the unligated veins to
overcome the occlusion of the blood flow in the treated retinas.
Our model based on the episcleral vein ligation model ex- This compensational increase of blood flow through the intact
hibits phenotypes that are essentially identical to that observed vein in response to the occlusion might enable retinal tissue to
by other models, and appears to recapitulate characteristic mor- escape from the ischemic damage. Moreover, we detected no
phological features of glaucoma. Correlated with chronic IOP change in the photoreceptors and the thickness of the outer nu-
elevation, our results showed a RGC loss and optic disc excava- clear layer between the treated and control retinas even at ad-
tion. Previous studies, using other glaucoma models, showed vanced stages of IOP elevation, which is consistent with the
that there was a 1.4e6% reduction of RGCs per week and ex- idea that there is no ischemic change in our model.
hibited significant cell loss immediately after IOP elevation Our analyses using TUNEL assays showed that RGC loss
(Garcia-Valenzuela et al., 1995; Laquis et al., 1998; Sawada was induced by apoptosis, consistent with previously reported
and Neufeld, 1999; Naskar et al., 2002; Ji et al., 2004). In con- glaucoma models (Morrison et al., 1997; Ishii et al., 2003;
trast, the loss of RGC in our ligation model is 1.1e1.5% per Fortune et al., 2004; Kanamori et al., 2004). TUNEL-positive
week and appears to progress more slowly compared to other cells were shown to be confined to retrogradely labeled RGCs
procedures. The late onset and less severe extent of RGC loss in whole-mount preparations, suggesting that RGC is selec-
most likely reflect the mild IOP elevation induced in our model. tively impaired during the chronic phases of IOP elevation
766 S. Yu et al. / Experimental Eye Research 83 (2006) 758e770

Fig. 7. Immunohistochemical analyses of retinal cells in sham-operated control retinas (A, C, E, G, I, K, M) and in glaucoma model retinas (B, D, F, H, J, L, N) at
32 weeks after operation, showing a decreased expression of p75 (A, B) and Brn3b (C, D) in the GCL in glaucoma retina. Calbindin (E, F), PKC (G, H) and Opsin
(I, J) showed no obvious difference between the glaucomatous retinas and controls. The expression of GFAP (K, L) was detected in a pattern that spans the retina
from INL to GCL in both the control and the glaucomatous retina. The expression of p27Kip1 (M, N) in the INL was decreased in glaucoma retinas. GCL, retinal
ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer. Scale bar: 50 mm.

in our model. Although there are several reports showing that markers of each retinal neuron as well as histological analyses
photoreceptors are also affected in glaucoma (Bayer et al., of retinal morphology showed that the retinal cells that were
2001; Grozdanic et al., 2003a), our immunohistochemical lost in this model were restricted to RGC, and there was no
analyses of the expression patterns of specific molecular sign of involvement of photoreceptors. Only Müller glial cells
showed a change in the marker expression. With many types
of insult to the eye, Müller cells were demonstrated to upregu-
late GFAP expression and also to dedifferentiate and prolifer-
ate at an early stage, and gradually reduced (Kim et al., 1998;
Wang et al., 2000; Bringmann and Reichenbach, 2001). Previ-
ous studies on glaucoma models also showed that GFAP ex-
pression is increased in Müller cells at least at early stages
of IOP elevation, but whether GFAP upregulation persists
into advanced stages remained obscure (Wang et al., 2000;
Thanos and Naskar, 2004; Woldemussie et al., 2004). In this
ligation glaucoma model, we focused on the advanced stage
of glaucoma retina. GFAP expression in Müller cells appeared
to be increased in the treated retina, but we could not detect
Fig. 8. Quantitative analyses of GFAP and p27Kip1 mRNAs at 30 weeks. Data a significant difference in GFAP mRNA expressions compared
are expressed as mean  S.E.M. GFAP expression did not exhibit statistically
significant difference, but the expression of p27kip1 showed statistically sig-
to control retinas. In contrast, p27Kip1, a molecule that plays
nificant decrease in the glaucoma retinas (Gla model) compared to the controls a critical role in exiting from the mitotic cell cycle (Dyer and
(Control). *P < 0.05. Cepko, 2001) in Müller cells, was significantly downregulated
S. Yu et al. / Experimental Eye Research 83 (2006) 758e770 767

Fig. 9. Immunohistochemical analyses of spatial and temporal patterns of neurotrophic factors, CNTF, bFGF, BDNF, and GDNF in the control (A, E, I, M) at
10 weeks and glaucomatous retina (Gla) at 10 weeks (B, F, J, N), 20 weeks (C, G, K, O) and 30 weeks (D, H, L, P). CNTF: AeD; bFGF: EeH; BDNF: IeL;
GDNF: MeP. See the details in the text. Note that the expression of CNTF in 10w glaucoma retina is upregulated compared to control retina. At advanced stage
of glaucoma (30 weeks after the treatment) all the factors including CNTF showed the decline in those expression. Sections of the retina were counterstained with
TO-PRO-3 (blue). GCL, retinal ganglion cell layer; INL, inner nuclear layer; ONL, outer nuclear layer. Scale bar: 50 mm.

in the treated retinas. Altogether, these results are partly loss observed in glaucoma might be the deprivation of neuro-
consistent with the idea that, even at advanced stage of glau- trophic factors that control the survival of RGCs (Johnson
comatous retina, Müller cells might be under control of dedif- et al., 1986; Ko et al., 2000). Indeed, applications of neurotro-
ferentiation and proliferation mechanism to work in the phic factors, including BDNF, CNTF, and GDNF, via various
regenerating process in response to IOP elevation. Further approaches has been shown to be beneficial for rescuing RGCs
studies are needed to see if reactive Müller cells might contrib- from death in glaucoma eyes (Naskar et al., 2000; Martin
ute to compensational repair of RGC loss in the glaucomatous et al., 2003; Ji et al., 2004). However, the spatial and
condition. temporal changes in expression patterns of neurotrophic fac-
tors by chronically elevated IOP are not fully understood
4.3. Neurotrophic factors expression in elevated IOP (Pease et al., 2000; Ji et al., 2004; Rudzinski et al., 2004)
and need to be examined to devise more efficient rescue
Neurotrophic factors play a central role in maintaining and approaches.
controlling the function of retinal cells in the developing and Our ligation model allowed us to analyze the expression
mature retina (Von Bartheld, 1998; Vecino et al., 2002). Previ- patterns of neurotrophic factors that might contribute to pre-
ous studies have suggested that one potential reason of RGC venting RGC loss. Immunohistochemical analyses for the
768 S. Yu et al. / Experimental Eye Research 83 (2006) 758e770

Fig. 10. Quantitative analyses of CNTF, bFGF, BDNF and GDNF mRNAs at 10, 20 and 30 weeks after the treatment. Datas are glaucoma retina/control retina ratio
at each point, and expressed as mean  S.E.M.(%). The expression level of CNTF and bFGF significantly decreased at 30 weeks, whereas that of BDNF and
GDNF did not change during the period of IOP elevation. *P < 0.05.

expression of growth factors at various time points revealed In conclusion, we reported here an animal model of glau-
a unique pattern for each of the factors examined. In the con- coma with episcleral vein ligation in rat eyes. Our analyses
trol retina, there was no obvious change in the expression of of this animal model have revealed that IOP was elevated to
growth factors throughout the follow-up time course (data a mild extent from the early postoperative stage and persisted
not shown). Previous studies showed that the expression of reproducibly for several months after the procedure. A gradual
BDNF in the retina was increased in the early glaucomatous increase of RGC-specific cell death preceding optic disc exca-
stage (Rudzinski et al., 2004). In our glaucoma model, how- vation was observed after several weeks of IOP elevation. Our
ever, we could not detect any increased expression of BDNF episcleral vein ligation model reiterated many critical charac-
in the GCL, possibly because of the difference in the timing teristics of human glaucoma and may, potentially, provide in-
of analyses. GDNF (Naskar et al., 2000) also did not show sights into the underlying molecular and cellular bases of
any increased expression during the chronic phase of IOP el- glaucomatous optic neuropathy.
evation. Another trophic factor, bFGF, that was normally pres-
ent in the GCL, INL, ONL and RPE cells (Connolly et al., Acknowledgements
1992), was shown to be upregulated in Müller cells by light
exposure and ischemia (Miyashiro et al., 1998; Walsh et al., The authors thank Yoshinaga Tochikawa of Fujimoto
2001). In contrast, in this ligation model, there was no appar- Pharmacentical Corporation for instructions in RGC labeling
ent upregulation of bFGF and the expression of bFGF declined and counting methods, and Mari Dezawa and Hiroto Ishikawa
in the late glaucoma stage. Among the growth factors exam- of Kyoto University for their advice on histological and immu-
ined by immunohistochemical analyses, CNTF expression nohistochemical analyses.
alone appeared to be increased especially in the INL at an
early glaucoma stage. A similar expression pattern of changes
in CNTF expression was also detected in the optic nerve tran- References
section model and NMDA- and kainic acid-induced neuronal
Bayer, A.U., Danias, J., Brodie, S., Maag, K.P., Chen, B., Shen, F.,
death (Honjo et al., 2000; Casson et al., 2004). Although we Podos, S.M., Mittag, T.W., 2001. Electroretinographic abnormalities in
could not detect increase in the expression of CNTF by real- a rat glaucoma model with chronic elevated intraocular pressure. Exp.
time PCR approach, these observations may, therefore, suggest Eye Res. 72, 667e677.
CNTF as a protective factor that is especially dedicated to the Bringmann, A., Reichenbach, A., 2001. Role of Muller cells in retinal degen-
loss of RGCs under several pathological circumstances (Chun erations. Front. Biosci. 6, E72eE92.
Casson, R.J., Chidlow, G., Wood, J.P., Vidal-Sanz, M., Osborne, N.N., 2004.
et al., 2000; Sarup et al., 2004). Taken together, the results of The effect of retinal ganglion cell injury on light-induced photoreceptor
our study on the expression patterns of neurotrophic factors es- degeneration. Invest. Ophthalmol. Vis. Sci. 45, 685e693.
pecially highlight the potential role of CNTF in the control of Chun, M.H., Ju, W.K., Kim, K.Y., Lee, M.Y., Hofmann, H.D., Kirsch, M.,
RGC survival during the early stage of IOP elevation. Further Oh, S.J., 2000. Upregulation of ciliary neurotrophic factor in reactive
analyses and identification of other trophic factors, which are Muller cells in the rat retina following optic nerve transection. Brain
Res. 868, 358e362.
spatially and temporally regulated during the course of IOP el- Connolly, S.E., Hjelmeland, L.M., LaVail, M.M., 1992. Immunohistochemical
evation, may provide potential insights into understanding way localization of basic fibroblast growth factor in mature and developing ret-
of reversing RGC defects in glaucoma. inas of normal and RCS rats. Curr. Eye Res. 11, 1005e1017.
S. Yu et al. / Experimental Eye Research 83 (2006) 758e770 769

Danias, J., Shen, F., Kavalarakis, M., Chen, B., Glodblum, D., Lee, K., factor as a protection: retinal ganglion cells in a rat glaucoma model. In-
Zamora, M.-F., Su, Y., Podos, M., Mittag, T., 2006. Characterization of ret- vest. Ophthalmol. Vis. Sci. 44, 4357e4365.
inal damage in the episcleral vein cauterization rat glaucoma model. Exp. Mermoud, A., Baerveldt, G., Minckler, D.S., Lee, M.B., Rao, N.A., 1994. Intra-
Eye Res. 82, 219e228. ocular pressure in Lewis rats. Invest. Ophthalmol. Vis. Sci. 35, 2455e2460.
Dyer, M.A., Cepko, C.L., 2001. p27Kip1 and p57Kip2 regulate proliferation in Mey, J., Thanos, S., 1993. Intravitreal injections of neurotrophic factors sup-
distinct retinal progenitor cell populations. J. Neurosci. 21, 4259e4271. port the survival of axotomized retinal ganglion cells in adult rats in
Fortune, B., Bui, B.V., Morrison, J.C., Johnson, E.C., Dong, J., Cepurna, W.O., vivo. Brain Res. 602, 304e317.
Jia, L., Barber, S., Cioffi, G.A., 2004. Selective ganglion cell functional Mittag, T.W., Danias, J., Pohorenec, G., Yuan, H.M., Burakgazi, E., Chalmers-
loss in rats with experimental glaucoma. Invest. Ophthalmol. Vis. Sci. Redman, R., Podos, S.M., Tatton, W.G., 2000. Retinal damage after 3 to 4
45, 1854e1862. months of elevated intraocular pressure in a rat glaucoma model. Invest.
Garcia-Valenzuela, E., Shareef, S., Walsh, J., Sharma, S.C., 1995. Pro- Ophthalmol. Vis. Sci. 41, 3451e3459.
grammed cell death of retinal ganglion cells during experimental glau- Miyashiro, M., Ogata, N., Takahashi, K., Matsushima, M., Yamamoto, C.,
coma. Exp. Eye Res. 61, 33e44. Yamada, H., Uyama, M., 1998. Expression of basic fibroblast growth fac-
Glovinsky, Y., Quigley, H.A., Dunkelberger, G.R., 1991. Retinal ganglion cell tor and its receptor mRNA in retinal tissue following ischemic injury in the
loss is size dependent in experimental glaucoma. Invest. Ophthalmol. Vis. rat. Graefes Arch. Clin. Exp. Ophthalmol. 236, 295e300.
Sci. 32, 484e491. Miyawaki, T., Uemura, A., Dezawa, M., Yu, R.T., Ide, C., Nishikawa, S.,
Grozdanic, S.D., Betts, D.M., Sakaguchi, D.S., Allbaugh, R.A., Kwon, Y.H., Honda, Y., Tanabe, Y., Tanabe, T., 2004. Tlx, an orphan nuclear receptor,
Kardon, R.H., 2003a. Laser-induced mouse model of chronic ocular hyper- regulates cell numbers and astrocyte development in the developing retina.
tension. Invest. Ophthalmol. Vis. Sci. 44, 4337e4346. J. Neurosci. 24, 8124e8134.
Grozdanic, S.D., Betts, D.M., Sakaguchi, D.S., Kwon, Y.H., Kardon, R.H., Morrison, J.C., Moore, C.G., Deppmeier, L.M., Gold, B.G., Meshul, C.K.,
Sonea, I.M., 2003b. Temporary elevation of the intraocular pressure by Johnson, E.C., 1997. A rat model of chronic pressure-induced optic nerve
cauterization of vortex and episcleral veins in rats causes functional defi- damage. Exp. Eye Res. 64, 85e96.
cits in the retina and optic nerve. Exp. Eye Res. 77, 27e33. Naskar, R., Vorwerk, C.K., Dreyer, E.B., 2000. Concurrent downregulation of
Honjo, M., Tanihara, H., Kido, N., Inatani, M., Okazaki, K., Honda, Y., 2000. a glutamate transporter and receptor in glaucoma. Invest. Ophthalmol. Vis.
Expression of ciliary neurotrophic factor activated by retinal Muller cells Sci. 41, 1940e1944.
in eyes with NMDA- and kainic acid-induced neuronal death. Invest. Oph- Naskar, R., Wissing, M., Thanos, S., 2002. Detection of early neuron degen-
thalmol. Vis. Sci. 41, 552e560. eration and accompanying microglial responses in the retina of a rat model
Ishii, Y., Kwong, J.M., Caprioli, J., 2003. Retinal ganglion cell protection with of glaucoma. Invest. Ophthalmol. Vis. Sci. 43, 2962e2968.
geranylgeranylacetone, a heat shock protein inducer, in a rat glaucoma Pease, M.E., McKinnon, S.J., Quigley, H.A., Kerrigan-Baumrind, L.A.,
model. Invest. Ophthalmol. Vis. Sci. 44, 1982e1992. Zack, D.J., 2000. Obstructed axonal transport of BDNF and its receptor
Ji, J.Z., Elyaman, W., Yip, H.K., Lee, V.W., Yick, L.W., Hugon, J., So, K.F., TrkB in experimental glaucoma. Invest. Ophthalmol. Vis. Sci. 41, 764e774.
2004. CNTF promotes survival of retinal ganglion cells after induction Pederson, J.E., Gaasterland, D.E., 1984. Laser-induced primate glaucoma. I.
of ocular hypertension in rats: the possible involvement of STAT3 pathway. Progression of cupping. Arch. Ophthalmol. 102, 1689e1692.
Eur. J. Neurosci. 19, 265e272. Quigley, H.A., Green, W.R., 1979. The histology of human glaucoma cupping
Jia, L., Cepurna, W.O., Johnson, E.C., Morrison, J.C., 2000. Effect of general and optic nerve damage: clinicopathologic correlation in 21 eyes. Ophthal-
anesthetics on IOP in rats with experimental aqueous outflow obstruction. mology 86, 1803e1830.
Invest. Ophthalmol. Vis. Sci. 41, 3415e3419. Quigley, H.A., Dunkelberger, G.R., Green, W.R., 1988. Chronic human glau-
John, S.W., Smith, R.S., Savinova, O.V., Hawes, N.L., Chang, B., Turnbull, D., coma causing selectively greater loss of large optic nerve fibers. Ophthal-
Davisson, M., Roderick, T.H., Heckenlively, J.R., 1998. Essential iris atro- mology 95, 357e363.
phy, pigment dispersion, and glaucoma in DBA/2J mice. Invest. Ophthal- Quigley, H.A., Nickells, R.W., Kerrigan, L.A., Pease, M.E., Thibault, D.J.,
mol. Vis. Sci. 39, 951e962. Zack, D.J., 1995. Retinal ganglion cell death in experimental glaucoma
Johnson, J.E., Barde, Y.A., Schwab, M., Thoenen, H., 1986. Brain-derived and after axotomy occurs by apoptosis. Invest. Ophthalmol. Vis. Sci. 36,
neurotrophic factor supports the survival of cultured rat retinal ganglion 774e786.
cells. J. Neurosci. 6, 3031e3038. Rudzinski, M., Wong, T.P., Saragovi, H.U., 2004. Changes in retinal expres-
Kanamori, A., Nakamura, M., Nakanishi, Y., Nagai, A., Mukuno, H., sion of neurotrophins and neurotrophin receptors induced by ocular hyper-
Yamada, Y., Negi, A., 2004. Akt is activated via insulin/IGF-1 receptor tension. J. Neurobiol. 58, 341e354.
in rat retina with episcleral vein cauterization. Brain Res. 1022, 195e204. Sapieha, P.S., Peltier, M., Rendahl, K.G., Manning, W.C., Di Polo, A., 2003.
Kim, I.B., Kim, K.Y., Joo, C.K., Lee, M.Y., Oh, S.J., Chung, J.W., Chun, M.H., Fibroblast growth factor-2 gene delivery stimulates axon growth by adult
1998. Reaction of Muller cells after increased intraocular pressure in the retinal ganglion cells after acute optic nerve injury. Mol. Cell. Neurosci.
rat retina. Exp. Brain Res. 121, 419e424. 24, 656e672.
Ko, M.L., Hu, D.N., Ritch, R., Sharma, S.C., 2000. The combined effect of Sarup, V., Patil, K., Sharma, S.C., 2004. Ciliary neurotrophic factor and its re-
brain-derived neurotrophic factor and a free radical scavenger in experi- ceptors are differentially expressed in the optic nerve transected adult rat
mental glaucoma. Invest. Ophthalmol. Vis. Sci. 41, 2967e2971. retina. Brain Res. 1013, 152e158.
Krupin, T., Feitl, M., Roshe, R., Lee, S., Becker, B., 1980. Halothane anesthe- Sawada, A., Neufeld, A.H., 1999. Confirmation of the rat model of chronic,
sia and aqueous humor dynamics in laboratory animals. Invest. Ophthal- moderately elevated intraocular pressure. Exp. Eye Res. 69, 525e531.
mol. Vis. Sci. 19, 518e521. Shareef, S.R., Garcia-Valenzuela, E., Salierno, A., Walsh, J., Sharma, S.C.,
Laquis, S., Chaudhary, P., Sharma, S.C., 1998. The patterns of retinal ganglion 1995. Chronic ocular hypertension following episcleral venous occlusion
cell death in hypertensive eyes. Brain Res. 784, 100e104. in rats. Exp. Eye Res. 61, 379e382.
Levkovitch-Verbin, H., Quigley, H.A., Martin, K.R., Valenta, D., Thanos, S., Naskar, R., 2004. Correlation between retinal ganglion cell death
Baumrind, L.A., Pease, M.E., 2002. Translimbal laser photocoagulation and chronically developing inherited glaucoma in a new rat mutant. Exp.
to the trabecular meshwork as a model of glaucoma in rats. Invest. Oph- Eye Res. 79, 119e129.
thalmol. Vis. Sci. 43, 402e410. Ueda, J., Sawaguchi, S., Hanyu, T., Yaoeda, K., Fukuchi, T., Abe, H.,
Mabuchi, F., Lindsey, J.D., Aihara, M., Mackey, M.R., Weinreb, R.N., 2004. Ozawa, H., 1998. Experimental glaucoma model in the rat induced by laser
Optic nerve damage in mice with a targeted type I collagen mutation. In- trabecular photocoagulation after an intracameral injection of India ink.
vest. Ophthalmol. Vis. Sci. 45, 1841e1845. Jpn. J. Ophthalmol. 42, 337e344.
Martin, K.R., Quigley, H.A., Zack, D.J., Levkovitch-Verbin, H., Vecino, E., Garcia-Grespo, D., Garcia, M., Martinez-Millan, L., Sharma, S.C.,
Kielczewski, J., Valenta, D., Baumrind, L., Pease, M.E., Klein, R.L., Carrascal, E., 2002. Rat retinal ganglion cells co-express brain derived
Hauswirth, W.W., 2003. Gene therapy with brain-derived neurotrophic neurotrophic factor (BDNF) and its receptor TrkB. Vision Res. 42, 151e157.
770 S. Yu et al. / Experimental Eye Research 83 (2006) 758e770

Vidal-Sanz, M., Villegas-Perez, M.P., Bray, G.M., Aguayo, A.J., 1988. Persis- Wang, X., Tay, S.S., Ng, Y.K., 2000. An immunohistochemical study of neu-
tent retrograde labeling of adult rat retinal ganglion cells with the carbo- ronal and glial cell reactions in retinae of rats with experimental glaucoma.
cyanine dye diI. Exp Neurol. 102, 92e101. Exp Brain Res. 132, 476e484.
Von Bartheld, C.S., 1998. Neurotrophins in the developing and regenerating Woldemussie, E., Wijono, M., Ruiz, G., 2004. Muller cell response to laser-
visual system. Histol. Histopathol. 13, 437e459. induced increase in intraocular pressure in rats. Glia 47, 109e119.
Walsh, N., Valter, K., Stone, J., 2001. Cellular and subcellular patterns of ex- Wu, W.C., Lai, C.C., Chen, S.L., Sun, M.H., Xiao, X., Chen, T.L., Tsai, R.J.,
pression of bFGF and CNTF in the normal and light stressed adult rat ret- Kuo, S.W., Tsao, Y.P., 2004. GDNF gene therapy attenuates retinal ische-
ina. Exp. Eye Res. 72, 495e501. mic injuries in rats. Mol. Vis. 10, 93e102.

You might also like