ArticlePDF Available

Age of Rats Seriously Affects the Degree of Retinal Damage Induced by Acute High Intraocular Pressure

Authors:

Abstract and Figures

Purpose: To investigate whether retinal impairment was affected by age of rats in acute glaucoma model. Methods: Young adult and aged rats were randomly divided into normal control, 45 mmHg, 60 mmHg and 90 mmHg groups. Intraocular pressures (IOP) of rats were acutely elevated to 45 mmHg, 60 mmHg and 90 mmHg, respectively. Neuron loss in ganglion cell layer (GCL) and activation of retinal macrolgia and microglia 3 days after high IOP treatment were detected by immunofluorescence and further quantitatively analyzed. Results: Compared with normal control, significant loss of neurons at GCL of young adult retina wasn't detected until IOP treatment of 90 mmHg. In contrast, obvious loss of neurons at GCL of aged retina was detected at IOP of 45 mmHg (p = 0.002 for central; p = 0.001 for peripheral). The loss level of neurons of aged retina was significantly higher than that of young adult retina at different IOP treatments. Compared with the young adult retina, high IOP induced more significant increase at area percentage of microglia and microglia number in inner part of aged retina. Activation of microglia and macroglia was either in parallel to or earlier than neuron loss of GCL of aged and young adult retina. Conclusion: Our data suggest there exists an age-related susceptibility of rat retina to the increased IOP. Therefore, the effect of ages should be considered at glaucoma study of rat models.
Content may be subject to copyright.
Current Eye Research, Early Online, 1–7, 2014
!Informa Healthcare USA, Inc.
ISSN: 0271-3683 print / 1460-2202 online
DOI: 10.3109/02713683.2014.922194
ORIGINAL ARTICLE
Age of Rats Seriously Affects the Degree of Retinal
Damage Induced by Acute High Intraocular Pressure
Chang Tan
1
,TuHu
2
, Ming-chao Peng
1
, Shu-li Liu
1
,
Jian-bin Tong
1
, Wen Ouyang
1
and Yuan Le
1
1
Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha, P.R. China,
2
Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University,
Changsha, P.R. China
ABSTRACT
Purpose: To investigate whether retinal impairment was affected by age of rats in acute glaucoma model.
Methods: Young adult and aged rats were randomly divided into normal control, 45 mmHg, 60 mmHg and
90 mmHg groups. Intraocular pressures (IOP) of rats were acutely elevated to 45 mmHg, 60 mmHg
and 90 mmHg, respectively. Neuron loss in ganglion cell layer (GCL) and activation of retinal macrolgia
and microglia 3 days after high IOP treatment were detected by immunofluorescence and further quantitatively
analyzed.
Results: Compared with normal control, significant loss of neurons at GCL of young adult retina wasn’t detected
until IOP treatment of 90 mmHg. In contrast, obvious loss of neurons at GCL of aged retina was detected at
IOP of 45 mmHg (p= 0.002 for central; p= 0.001 for peripheral). The loss level of neurons of aged retina was
significantly higher than that of young adult retina at different IOP treatments. Compared with the young adult
retina, high IOP induced more significant increase at area percentage of microglia and microglia number
in inner part of aged retina. Activation of microglia and macroglia was either in parallel to or earlier than
neuron loss of GCL of aged and young adult retina.
Conclusion: Our data suggest there exists an age–related susceptibility of rat retina to the increased IOP.
Therefore, the effect of ages should be considered at glaucoma study of rat models.
Keywords: Aging, glaucoma, intraocular pressures, retina, rat
INTRODUCTION
Glaucoma is a common age-related eye disease that
affects 3% of the worldwide population over the age
of 40, making it the second-leading cause of blind-
ness.
1–3
It is characterized by the visual field loss,
the death of a substantial number of retinal ganglion
cells (RGCs) and the loss of their axons in the optic
nerve.
4
Lots of clinical studies have shown that age
4
and elevated intraocular pressures (IOP)
4,5
are the
most extensive document risk factors of glaucoma.
By now, IOP reduction is the only treatment strategy
for all types of glaucoma.
4
In order to investigate
the neuropathologic mechanism of glaucoma and
develop effective therapeutic interventions, many
kinds of experimental glaucoma models were
formed by elevating IOP or changing gene MYOC or
a1 subunit of collagen type 1 in rats,
6,7
mice,
8,9
monkeys,
10,11
dogs,
12
cats,
12
and several other spe-
cies.
12
Studies based on these animal models have
shown that neuropathology is related with depriv-
ation of target neurotrophic factor,
13,14
oxidative
stress,
15,16
mitochondrial dysfunction,
17
excitotoxic
damage,
18–20
and inflammation,
21
activation of apop-
totic signals,
22,23
ischemia.
24,25
Unfortunately, the
pathophysiologic mechanisms underlying glaucoma
are not understood, though researches in the field
of glaucoma are substantial. In addition, in these
Correspondence: Yuan Le, Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha, P.R. China.
E-mail: leyuanxy@csu.edu.cn
Received 18 December 2013; revised 21 April 2014; accepted 1 May 2014; published online 25 August 2014
1
Curr Eye Res Downloaded from informahealthcare.com by Central South University on 09/18/14
For personal use only.
animal models of glaucoma, only young adult ani-
mals were used, neglecting the fact that glaucoma
is an age-related eye disorder and that age is an
important risk factor of glaucoma. Interestingly,
Samuel et al. found that RGCs density and synapse
density of inner plexiform layer and the areas of
dendritic and axonal arbors of RGCs all decreased at
the aged retina, compared to that of the young adult
retina.
26
Moreover, Wang et al. found that the loss
of RGCs after optic nerve crush in old mice was faster
than that in young mice.
27
Bakalash et al. found that
the protection of chondroitin sulfate-derived disac-
charide on retinal cells from IOP-induced death was
age-dependent in rats.
28
These data indicate that age
significantly impacts on the retinal injury and recov-
ery. On the other hand, high IOP is the most extensive
document risk factors of glaucoma. Kawai et al. also
detected that acute high IOP treatment induced
significant loss of RGCs at aged retinae that were
pretreated with chronic high IOP, compared to that
of age-matched rats.
29
However, in this study, the rats
were pretreated with chronic high IOP, therefore it
remains unknown whether aged and young adult rats
with same IOP treatment would yield the same retinal
injury.
Rat is a commonly-used model animal of glau-
coma. Rat shares similar anatomical
30,31
and develop-
mental
32,33
features of the anterior chamber, and
similar aqueous outflow pathway with the human.
Rat and human genomes are highly conservative.
34
These provide the basis for the use of rats as
a glaucoma model animal. In addition, rats are
inexpensive and easy to house and handle.
35
Their
eyes are easy to obtain, and the sample number for
studies can be large.
35
Moreover, elevating IOP of
rats reproduced the core phenotypes of glaucoma,
including RGC loss and damage of the optic nerve.
36
Thus, rat is a good model animal of glaucoma. Here
we constructed the rat glaucoma model by elevating
the IOP with cannulation of the anterior chamber,
and compared the retinal difference between young
adult and aged rats 3 d after IOP treatment of
45 mmHg, 60 mmHg or 90 mmHg. We found that
increased IOP induced more serious retinal damage
at aged retina than at young adult retina. At the same
time, increased IOP also activated more the astrocytes
and microglia at the aged retina.
MATERIALS AND METHODS
Animals and Grouping
Twelve young adult (aged 2 months, 200–250 g) and
twelve aged (aged 18 months, 500–550 g) female
Sprague-Dawley rats were purchased from Central
South University (P.R. China). All rats were raised
under controlled environmental conditions on a 12 h
light/dark cycle with ad libitum access to food and
water. All experimental protocols were approved by
the local animal ethics committee, and the guidelines
for animal experiments of Central South University
Young adult and aged rats were randomly divided
into normal control (n= 3, 6 eyes), 45 mmHg (n=3,
6 eyes), 60 mmHg (n= 3, 6 eyes), 90 mmHg (n=3,
6 eyes) groups. Intraocular pressures of rats in
45 mmHg, 60 mmHg, 90 mmHg (n= 3, 6 eyes) groups
were increased acutely to 45 mmHg, 60 mmHg and
90 mmHg, respectively. All the rats were killed at
3 days after high intraocular pressures.
Acute IOP Model
The animal model was prepared following reported
method.
35
Briefly, under anesthesia of 2% pentobar-
bital (40 mg/kg), 30-gauge needle connected to the
instillation instrument filled with normal saline
were inserted into the anterior chamber of rats. The
intraocular pressures was elevated to 45 mmHg,
60 mmHg or 90 mmHg, and then maintained for
60 min. After maintenance of 60 min, the 90 mmHg
of intraocular pressures was decreased through
80 mmHg for 5 min, 70 mmHg for 5 min, 60 mmHg
for 5 min, 30 mmHg for 5 min. finally, the needle
inserted into the anterior chamber was taken out.
For the condition of 45 mmHg and 60 mmHg of
intraocular pressures, after maintenance of 60 min,
the intraocular pressure was directly decreased
to 30 mmHg. Five minutes later, the needle inserted
into the anterior chamber was taken out.
Retinal Tissue Preparation
Under deep anesthesia, rats were firstly infused with
0.9% saline at 37 C, and then with 4% paraformalde-
hyde. The eyes were enucleated. After removing the
corneas and the lenses, the remainder of the eye
including the retina was post-fixed in 4% paraformal-
dehyde for 4 hr more at 4 C and then immersed in
30% sucrose. Cross sections were cut in parallel to the
equator of eyes through the optic disc of the retinae
at a thickness of 14 mm by a cryostat machine,
mounted on glass slides, dried at room temperature,
and finally stored at 20 C.
Immunofluorescence
Retinal sections stored at 20 C were taken out and
warmed at room temperature. After washing with
0.01 M phosphate buffered saline (PBS) for 10 min,
sections were incubated in blocking solution (5% BSA
and 0.3% Triton X- 100 in 0.1 M PB) for 1 hr at room
temperature. Then the sections were incubated in
2C. Tan et al.
Current Eye Research
Curr Eye Res Downloaded from informahealthcare.com by Central South University on 09/18/14
For personal use only.
primary antibodies
36–38
(NeuN, 1:1000, Millipore,
Billerica, MA; Iba-1, 1:1000, Wako Chemical,
Richmond, MA; GFAP, 1:2000, Millipore) overnight
at 4 C. On the second day, these sections were
washed with 0.01 M PBS for three times and then
incubated in the secondary antibodies labeled with
fluorescent dyes (1:200, Jackson Immuno research) for
2 hours at room temperature. Through three washes
of PBS, these sections were covered with mounting
medium with DAPI (vector). As negative controls, an
adjacent series of sections were processed using the
same procedures without the primary antibodies.
Analysis
Three sections from each eye for each staining were
chosen. 1/6, 3/6, and 5/6 of the retinal radius away
from the optic nerve head were referred to as central,
middle, and peripheral retina, respectively
39
. Images
of central (about 600 mm to the midpoint of optic nerve
head) and peripheral(about 4000 mm to the midpoint
of optic nerve head) parts of retinal sections were
captured under 40Objective lens (image field:
430 mm320 mm) on a confocal microscopy of Leica.
The neuron of retinal ganglion cell layers was defined
by co-labeling of NeuN (neuron marker) and DAPI
(nuclear marker). The microglia was defined by co-
labeling of Iba1 (microglia marker) and DAPI (nuclear
marker). The number of neuron and microglia in
retinal ganglion cell layers of captured pictures were
blindly counted, respectively. Percentage of Iba1
positive area to area of inner retina (including nerve
fiber layer, ganglion cell layer and inner plexiform
layer) and relative mean gray value of GFAP staining
at inner retina were measured by Image J.
40,41
For
quantitaion of each test, 36 images were used.
All data are presented as mean ± standard devi-
ation (mean ± SD). Two-way analysis of variance
(ANOVA) followed by LSD test were used for
means comparisons. PValues50.05 were considered
statistically significant.
RESULTS
Same High IOP Resulted in More Serious
Loss of Neurons at the Ganglion Cell Layer
of Aged Retinae than at Young Adult Retinae
RGC loss is the main pathological feature of glaucoma
retina. Thus we detected the neuron loss of ganglion
cell layer (GCL) of aged and young adult retina
3 days after IOP treatment of 45 mmHg, 60 mmHg,
and 90 mmHg (Figure 1). Two way ANOVA analysis
showed that aging significantly increased neuron loss
of GCL 3 days after IOP treatment (F(1,40)=138.99,
p50.001, for central; F(1,40)=84.14, p50.001, for
peripheral). Moreover, IOP treatment also
exhibited significant effects on neuron loss of GCL
(F (3,40)=214.13, p50.001, for central; F(3,40)=109.10,
p50.001, for peripheral). Further analysis showed
that at the normal control, the number of neurons
in GCL of peripheral (10.16 ± 0.87) and central
(15.33 ± 1.03) aged retina were not different from that
of young adult retina, respectively (p= 0.220 for
central; p= 0.341 for peripheral) (Figure 1). At IOP
treatment of 45 mmHg, the number of neurons in GCL
of peripheral (6.16 ± 0.97) and central (13.00 ± 1.11)
aged retina were significantly less than that of normal
aged retina, respectively (p= 0.002 for central; p= 0.001
for peripheral) (Figure 1). More obvious neuron loss
of peripheral and central aged retina was detected at
IOP treatment of 60 mmHg and 90 mmHg. However,
significant neuron loss of peripheral (3.00 ± 0.35) and
central (7.16 ± 1.17) retina could be detected at IOP
treatment of 90 mmHg at the young adult retinae,
compared to that of normal young adult retinae
(p50.001 for central; p50.001 for peripheral)
(Figure 1). These data implied that aged retina was
susceptible to IOP, compared to the young adult
retina.
Microglia of Aged Retinae was activated
more easily than that of Young Adult
Retinae under High IOP Treatment
Activated microglia played important roles in damage
of glaucoma by releasing inflammatory factors.
Previous results showed that peripheral retina was
susceptible to changed IOP, compared to the central
retina.
39
Thus we detected the activation of microglia
of peripheral retina by counting the number and the
area percentage of microglia marked by Iba1 in inner
retina (Figure 2). Two-way ANOVA analysis showed
that aging significantly increased the activation of
microglia 3 days after IOP treatment (F (1,40)=116.72,
p50.001, for area percentage; F (1,40)=156.89,
p50.001, for microglia number). Moreover, IOP treat-
ment also significantly affected the activation of
microglia (F (3,40)=250.13, p50.001, for area percent-
age; F(3,40)=190.03,p50.001, for microglia number)
(Figure 2). We found that at the normal control, the
number(1.00 ± 0.58) and the area percentage
(4.72% ± 0.51%) of microglia of aged retinae were not
different from that of young adult retina(p= 0.088
for number; p= 0.051 for area percetage) (Figure 2).
At day 3 after IOP treatments of 45 mmHg, 60 mmHg,
or 90 mmHg, the number and the area percentage of
microglia at aged and young adult retina both were
significantly increased, compared to that of normal
control(p50.05) (Figure 2). The number and the area
percentage of microglia of aged retinae were signifi-
cantly higher than that of young adult retina at each
IOP treatment (p50.05) (Figure 2). These showed that
Age of Rats Affects the Degree of Retinal Damage 3
!2014 Informa Healthcare USA, Inc.
Curr Eye Res Downloaded from informahealthcare.com by Central South University on 09/18/14
For personal use only.
increased IOP induced more obvious activation of
microglia at aged retina than at young adult retina.
Macroglia was Activated at the Retinae of
Aged and Young Adult Retina Under High
IOP Treatment
Mu
¨ller glial cells and astrocytes belong to macroglia in
the retina. Activation of macroglia was characterized
by increased processes and enlarged cell body. Here
we detected macroglia activation by measurement of
the relative gray value of GFAP staining, the marker
of macroglia, in the inner retina. Similar to that in
microglia, increased IOP induced stronger GFAP
expression at ganglion cell layer and more obvious
GFAP positive processes in inner plexiform layer at
aged and young adult retina, compared with the
normal retina (Figure 3). It was noticed that at aged
retina, many GFAP positive processes extended into
the inner plexiform layer, even into the inner nuclear
layer after high IOP treatment (Figure 3).
Two-way ANOVA analysis of relative mean gray
value of GFAP staining at the inner retina showed that
age didn’t significantly increased the activation of
macroglia 3 days after IOP treatment (F (1,40)=3.24,
FIGURE 1 Neurons of ganglion cell layer of aged retina were more susceptible to IOP damage than that of young adult retina.
Neurons of ganglion cell layer were marked by NeuN (red). Compared with the age-matched normal control (A, E, I, M), IOP
treatment of 45 mmHg, 60 mmHg, and 90 mmHg induced the loss of neurons at ganglion cell layer of young adult (B–D, J–L) and aged
(F–H, N–P) retinae 3 days after treatment. Quantitative analysis showed that at central (F) and peripheral (N) parts of aged retina, IOP
treatment of 45 mmHg was enough to induce significant loss of neurons of ganglion cell layer, compared to that of normal aged retina
(p= 0.002 for central; p= 0.001 for peripheral) (Q, R). In contrast, significant loss of neurons at ganglion cell layer of central and
peripheral young adult retinae was detected until IOP treatment of 90 mmHg (p50.001 for central; p50.001 for peripheral; Q, R).
GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner nuclear layer; ONL, outer nuclear layer. *p50.05 versus control;
#p50.05 versus matched part of young adult retina at the same IOP treatment. Bar = 50 mm.
4C. Tan et al.
Current Eye Research
Curr Eye Res Downloaded from informahealthcare.com by Central South University on 09/18/14
For personal use only.
p= 0.080). However, IOP treatment significantly
affected the activation of macroglia (F (3,40)=73.22,
p50.001; Figure 3). The relative mean gray value of
GFAP staining at the inner retina was significantly
increased at young adult and aged retinae, compared
with that of the normal control (p50.05; Figure 3).
The same IOP treatment didn’t produce significant
difference of relative mean gray value of GFAP
staining in the retina between aged and young adult
rats (p= 0.236 for 45 mmHg, p= 0.052 for 60 mmHg,
p= 0.561 for 90 mmHg; Figure 3).
DISCUSSION
The aim of this study was to investigate whether
retinal damage was affected by age of rats in
glaucoma model. We found that compared to that of
the young adult retina, (1) neurons at the ganglion cell
layer of aged retina were more susceptible to
increased IOP; (2) microglia of aged retina was more
easily activated by the increased IOP. These results
suggest that age of rats should be taken into account
at studying of experimental glaucoma.
Loss of RGCs was the main pathological feature of
glaucoma retina and the basis of visual defects.
42,43
Thus we first detected the loss of neuron at GCL after
different IOP treatments. We found that at young
adult retina, significant loss of neurons at GCL wasn’t
detected until IOP of 90 mmHg. However significant
loss of neurons at GCL of aged retina was detected
at IOP of 45 mmHg and more as IOP increased.
In addition, the loss of neurons in aged retina was
significantly higher than that of young adult retina at
different IOP treatments. These results suggested
that neurons of GCL of aged retina were more
susceptible to increased IOP, compared to young
adult retina. Our data are consistent with a recent
report in which after induction of prolonged cere-
bral hypoperfusion, 8-month-old mice showed more
severe white matter injury and working memory
dysfunction, compared with that of 2-month mice.
41
Previous researches showed that activation
of glial cells contributed much to the loss
of RGCs of glaucoma retina.
38–42
Inman et al.
found that in chronic glaucoma model of mice,
microglial cell number increased by two–fold.
44
Moreover, minocycline treatment reduced the retina
microglial activation in the DBA/2 J mouse model of
glaucoma, corresponding to the improved optic nerve
integrity.
45
Interestingly, in our study, we detected that
high IOP increased area percentage and number of
FIGURE 2 Activation of microglia at aged retina was more obvious than that of young adult retina 3 days after high IOP treatment.
Area percentage and number of microglia (green) were used to show the activation of microglia. Three days after IOP treatment of
45 mmHg, 60 mmHg, and 90 mmHg, microglia of inner retina of young adult (B–D) and aged (F–H) retinae both were activated,
compared with the age-matched normal control (A, E). Quantitative analysis showed that area percentage (J) and number (I) of
microglia of aged inner retina increased more, relative to that of young adult retina at each IOP treatment (p50.05). GCL, ganglion cell
layer; IPL, inner plexiform layer; INL, inner nuclear layer; ONL, outer nuclear layer. *p50.05 versus control; #p50.05 versus young
adult retina at the same IOP treatment. Bar = 50 mm.
Age of Rats Affects the Degree of Retinal Damage 5
!2014 Informa Healthcare USA, Inc.
Curr Eye Res Downloaded from informahealthcare.com by Central South University on 09/18/14
For personal use only.
microglia in inner retina at IOP of from 45 mmHg to
90 mmHg. Under the same IOP, the increased degrees
of area percentage of microglia and microglia number
at the inner part of aged retina were much higher than
that at young adult retina. Moreover, these changes of
microglia were in parallel to or earlier than the loss
of neurons at GCL. These suggested that more obvious
activation of microglia possibly contributed to the
easier loss of neurons at aged retinae after high IOP
treatment, compared to young adult retinae. But the
molecular mechanism that microglia differentially
contributed to the age-related susceptibility of neurons
of GCL needs to be further studied. In brief, we found
that age seriously affects the degree of retinal damage
induced by acute high intraocular pressure.
DECLARATION OF INTEREST
There is no conflict of interest. The authors alone are
responsible for the content and writing of the paper.
REFERENCES
1. Quigley HA, Broman AT. The number of people with
glaucoma worldwide in 2010 and 2020. Br J Ophthalmol
2006;90:262–267.
2. Chae B, Cakiner-Egilmez T, Desai M. Glaucoma medica-
tions. Insight 2013;38:5–9.
3. Voleti VB, Hubschman JP. Age-related eye disease.
Maturitas 2013;75:29–33.
4. Casson RJ, Chidlow G, Wood JP, Crowston JG, Goldberg I.
Definition of glaucoma:clinical and experimental concepts.
Clin Experiment Ophthalmol 2012;40:341–349.
5. Chen SD, Wang L, Zhang XL. Neuroprotection in glau-
coma: present and future. Chin Med J (Engl) 2013;126:
1567–1577.
6. Nitta E, Hirooka K, Tenkumo K, Fujita T, Nishiyama A,
Nakamura T, et al. Aldosterone: a mediator of retinal
ganglion cell death and the potential role in the pathogen-
esis in normal-tension glaucoma. Cell Death Dis 2013;4:
e711.
7. Sandalon S, Ko
¨nnecke B, Levkovitch-Verbin H, Simons M,
Hein K, Sa
¨ttler MB, et al. Functional and structural
evaluation of lamotrigine treatment in rat models of
acute and chronic ocular hypertension. Exp Eye Res 2013;
115:47–56.
8. Cone-Kimball E, Nguyen C, Oglesby EN, Pease ME,
Steinhart MR, Quigley HA. Scleral structural alterations
associated with chronic experimental intraocular pressure
elevation in mice. Mol Vis 2013;19:2023–2039.
9. Feng L, Chen H, Suyeoka G, Liu X. A laser-induced mouse
model of chronic ocular hypertension to characterize visual
defects. J Vis Exp 2013;(78).
10. Deng S, Wang M, Yan Z, Tian Z, Chen H, Yang X, et al.
Autophagy in retinal ganglion cells in a rhesus monkey
chronic hypertensive glaucoma model. PLoS One 2013;8:
e77100.
11. Cull G, Burgoyne CF, Fortune B, Wang L. Longitudinal
hemodynamic changes within the optic nerve head in
FIGURE 3 Macroglia was activated at the retinae of aged and young adult retina under high IOP treatment. Compared with the
normal control, increased IOP induced stronger GFAP staining (green) at ganglion cell layer and more obvious GFAP positive
processes in inner plexiform layer at young adult (B–D) and aged (F–H) retina 3 days after IOP treatment of 45 mmHg, 60 mmHg, and
90 mmHg. The relative mean gray value of GFAP staining at the inner retinae was significantly increased at young adult and aged
retinae, compared to that of the normal control (p50.05) (I). but no significant difference of relative mean gray value of GFAP staining
between aged and young adult retinae was founded (p40.05) (I). GCL, ganglion cell layer; IPL, inner plexiform layer; INL, inner
nuclear layer; ONL, outer nuclear layer. *p50.05 versus control; Bar = 50 mm.
6C. Tan et al.
Current Eye Research
Curr Eye Res Downloaded from informahealthcare.com by Central South University on 09/18/14
For personal use only.
experimental glaucoma. Invest Ophthalmol Vis Sci 2013;54:
4271–4277.
12. Bouhenni RA, Dunmire J, Sewell A, Edward DP. Animal
models of glaucoma. J Biomed Biotechnol 2012;2012:
692609.
13. Martin KR, Quigley HA, Zack DJ, Levkovitch-Verbin H,
Kielczewski J, Valenta D, et al. Gene therapy with
brain-derived neurotrophic factor as a protection: retinal
ganglion cells in a rat glaucoma model. Invest Ophthalmol
Vis Sci 2003;44:4357–4365.
14. Chen H, Weber AJ. BDNF enhances retinal ganglion
cell survival in cats with optic nerve damage. Invest
Ophthalmol Vis Sci 2001;42:966–974.
15. Kanamori A, Catrinescu MM, Kanamori N, Mears KA,
Beaubien R, Levin LA. Superoxide is an associated signal
for apoptosis in axonal injury. Brain 2010;133:2612–2625.
16. Moreno MC, Campanelli J, Sande P, Sanez DA, Keller SMI,
Rosenstein RE. Retinal oxidative stress induced by high
intraocular pressure. Free Radic Biol Med 2004;37:803–812.
17. Baltan S, Inman DM, Danilov CA, Morrison RS, Calkins DJ,
Horner PJ. Metabolic vulnerability disposes retinal gan-
glion cell axons to dysfunction in a model of glaucomatous
degeneration. J Neurosci 2010;30:5644–5652.
18. Seki M, Lipton SA. Targeting excitotoxic/free radical
signaling pathways for therapeutic intervention in glau-
coma. Prog Brain Res 2008;173:495–510.
19. Chuettauf F, Stein T, Choragiewicz TJ, Rejdak R, Bolz S,
Zurakowski D, et al. Caspase inhibitors protect
against NMDA-mediated retinal ganglion cell death. Clin
Experiment Ophthalmol 2011;39:545–554.
20. Dong CJ, Guo Y, Agey P, Wheeler L, Hare WA. Alpha2
adrenergic modulation of NMDA receptor function as a
major mechanism of RGC protection in experimental
glaucoma and retinal excitotoxicity. Invest Ophthalmol
Vis Sci 2008;49:4515–4522.
21. Tezel G. Immune regulation toward immunomodulation
for neuroprotection in glaucoma. Curr Opin Pharmacol
2013;13:23–31.
22. Libby RT, Li Y, Savinova OV, Barter J, Smith RS,
Nickells RW, et al. Susceptibility to neurodegeneration in
glaucoma is modified by Bax gene dosage. PLoS Genet
2005;1:17–26.
23. Li Y, Schlamp CL, Poulsen KP, Nickells RW.
Bax-dependent and independent pathways of retinal gan-
glion cell death induced by different damaging stimuli.
Exp Eye Res 2000;71:209–213.
24. Hafez AS, Bizzarro RL, Lesk MR. Evaluation of optic nerve
head and peripapillary retinal blood flow in glaucomapa-
tients, ocular hypertensives, and normal subjects. Am
J Ophthalmol 2003;136:1022–1031.
25. Choritz L, Machert M, Thieme H. Correlation of endothe-
lin-1 concentration in aqueous humor with intraocular-
pressure in primary open angle and pseudoexfoliation
glaucoma. Invest Ophthalmol Vis Sci 2012;53:7336–7342.
26. Samuel MA, Zhang Y, Meister M, Sanes JR. Age-related
alterations in neurons of the mouse retina. J Neurosci 2011;
31:16033–16044.
27. Wang AL1, Yuan M, Neufeld AH. Age-related changes
in neuronal susceptibility to damage:comparison of the
retinal ganglion cells of young and old mice before and
after optic nerve crush. Ann N Y Acad Sci 2007;1097:64–66.
28. Bakalash S, Rolls A, Lider O, Schwartz M. Chondroitin
sulfate-derived disaccharide protects retinal cells from
elevated intraocular pressure in aged and immunocom-
promised rats. Invest Ophthalmol Vis Sci 2007;48:
1181–1190.
29. Kawai SI, Vora S, Das S, Gachie E, Becker B, Neufeld AH.
Modeling of risk factors for the degeneration of retinal
ganglion cells after ischemia/reperfusion in rats:effects
of age, caloric restriction, diabetes, pigmentation, and
glaucoma. Faseb J. 2001;15:1285–1287.
30. van der Zypen E. Experimental morphological study on
structure and function of the filtration angle of the rat eye.
Ophthalmologica 1977;174:285–298.
31. Daimon T, Kazama M, Miyajima Y, Nakano M.
Immunocytochemical localization of thrombomodulin in
the aqueous humor passage of the rat eye. Histochem Cell
Biol 1997;108:121–131.
32. Reme C, Aeberhard B, Urner U. The development of the
chamber angle in the rat eye. Morphological characteristics
of developmental stages. Clin Experiment Ophthalmol
1983;220:139–153.
33. Nucci P, Tredici G, Manitto MP, Pizzini G, Brancato R.
Neuron-specific enolase and embryology of the trabecular
meshwork of the rat eye: an immunohistochemical study.
Int J Biol Markers 1992;7:253–255.
34. Bouhenni RA, Dunmire J, Sewell A, Edward DP. Animal
models of glaucoma. J Biomed Biotechnol 2012;2012:
692609.
35. Johnson TV, Tomarev SI. Rodent models of glaucoma.
Brain Res Bull 2010;81:349–358.
36. Joachim SC, Gramlich OW, Laspas P, Schmid H, Beck S,
von Pein HD, et al. Retinal ganglion cell loss is
accompanied by antibody depositions and increased
levels of microglia after immunization with retinal anti-
gens. PLoS One 2012;7:e40616.
37. Huang JF, Shang L, Zhang MQ, Wang H, Chen D, Tong JB,
et al. Differential neuronal expression of receptor interact-
ing protein 3 in rat retina: involvement in ischemic stress
response. BMC Neurosci 2013;14:16.
38. Huang J, Zhou L, Wang H, Luo J, Zeng L, Xiong K, Chen D.
Distribution of thrombospondins and their neur-
onal receptor a2d1 in the rat retina. Exp Eye Res 2013;111:
36–49.
39. Tong JB, Chen D, Zeng LP, Mo XY, Wang H, Huang JF, et al.
Differential changes of local blood supply in rat retinae are
involved in the selective loss of retinal ganglion cells
following the acute high intraocular pressure. Curr Eye
Res 2010;35:425–434.
40. Horstmann L, Schmid H, Heinen AP, Kurschus FC,
Dick HB, Joachim SC. Inflammatory demyelination
induces glia alterations and ganglion cell loss in the
retina of an experimental autoimmune encephalomyelitis
model. J Neuroinflammation 2013;10:120.
41. Joachim SC, Gramlich OW, Laspas P, Schmid H, Beck S,
von Pein HD, et al. Retinal ganglion cell loss is
accompanied by antibody depositions and increased
levels of microglia after immunization with retinal anti-
gens. PLoS One 2012;7:e40616.
42. Akhter N, Nix M, Abdul Y, Singh S, Husain S. Delta-opioid
receptors attenuate TNF-a-induced MMP-2 secretion from
human ONH astrocytes. Invest Ophthalmol Vis Sci 2013;
54:6605–6611.
43. Kernt M, Arend N, Buerger A, Mann T, Haritoglou C,
Ulbig MW, et al. Idebenone prevents human optic nerve
head astrocytes from oxidative stress, apoptosis, and
senescence by stabilizing BAX/Bcl-2 ratio. J Glaucoma
2013;22:404–412.
44. Inman DM, Horner PJ. Reactive non-proliferative gliosis
predominates in a chronic mouse model of glaucoma.
Glia 2007;55:942–953.
45. Bosco A, Inman DM, Steele MR, Wu G, Soto I,
Marsh-Armstrong N, et al. Reduced retina microglial
activation and improved optic nerve integrity with mino-
cycline treatment in the DBA/2J mouse model of glau-
coma. Invest Ophthalmol Vis Sci 2008;49:1437–1446.
Age of Rats Affects the Degree of Retinal Damage 7
!2014 Informa Healthcare USA, Inc.
Curr Eye Res Downloaded from informahealthcare.com by Central South University on 09/18/14
For personal use only.
... Consistent with these findings, we previously identified the existence of the age-related susceptibility of rat retinae to increased IOP [9]. Other reports suggested that agerelated mitochondria dysfunction and accumulation of oxidative damage make RGCs more vulnerable to damage in the progression of glaucomatous neurodegeneration [10,11]. ...
... Compared to the resting microglia and astrocytes, these "primed" microglia and astrocytes were more sensitive to inflammatory cues and displayed the more robust inflammatory response, as the higher expression of proinflammatory cytokines/chemokines and complement component [23,24]. Our previous results showed that high IOP treatment induces the more evident activation of microglia/macrophages (characterized by the more IBA1-(ionized calcium-binding adapter molecule 1-) positive cells and IBA1-positive stained area) in aged retinae than young adult retinae [9]. However, the proinflammatory features of microglia/macrophages have not been identified in this scenario. ...
... In the present study, we detected the formation of proinflammatory microglia/macrophages and neurotoxic astrocytes, deposition of complement C3, and production of proinflammatory cytokines (TNF and IL-1β) in the rat glaucoma model we previously used [9]. These findings raised the possibility that retinal neuroinflammatory response may contribute to the age-related vulnerability of RGCs in glaucoma. ...
Article
Full-text available
Purpose: To investigate whether retinal neuroinflammatory response was affected by aging in a rat model of acute glaucoma. Methods: Young adult and aged rats were randomly assigned into normal control, 45 mmHg, 60 mmHg, and 90 mmHg groups. Intraocular pressure (IOP) of rats was acutely elevated to 45 mmHg, 60 mmHg, and 90 mmHg, respectively. Three days after high IOP treatment, loss of retinal ganglion cells (RGCs), formation of proinflammatory microglia/macrophages and neurotoxic astrocytes, and deposition of complement C3 in the retina were detected by immunofluorescence. ELISA was used to assess the protein levels of proinflammatory cytokines TNF and IL-1β in the retina. Results: Compared with young adult retinae, (1) loss of RGCs was more severe in aged retinae under the same IOP treatment, (2) microglia/macrophages were more prone to adopt proinflammatory phenotype in aged retinae in response to elevated IOP, (3) high IOP treatment induced astrogliosis, formation of neurotoxic astrocytes, and deposition of complement C3 more easily in aged retinae, and (4) aged retinae induced higher levels of proinflammatory cytokines TNF and IL-1β under the same IOP treatment. Conclusion: Our data indicated that aging affects the degree of retinal neuroinflammatory response initiated by ocular hypertension, which may contribute to the age-related susceptibility of RGCs to elevated IOP.
... In this OHT animal model, mainly young animals were used. Considering that glaucoma is an age-related pathology, and that age is the main risk factor, most of the studies should have been performed on aged animals, which is not the case due to animal maintenance difficulties (Tan et al., 2015). An acute elevation of IOP in a rat model has shown a greater activation of macroglial and microglial cells in aged mice than in young adult mice (Tan et al., 2015). ...
... Considering that glaucoma is an age-related pathology, and that age is the main risk factor, most of the studies should have been performed on aged animals, which is not the case due to animal maintenance difficulties (Tan et al., 2015). An acute elevation of IOP in a rat model has shown a greater activation of macroglial and microglial cells in aged mice than in young adult mice (Tan et al., 2015). In addition to an acute IOP elevation (50 mmHg for 30 min) in aged mice, a significantly reduction was observed in the function of the inner retina . ...
... The differences between these results and those found in Damani's study (Damani et al., 2011) could be due to the fact that they studied mice older than ours (18-24 months), since our 15-month-old mice are in the early stages of aging, and the changes we observed in this study could be the first changes that microglial cells undergo in the aging process. In a rat model, Tan et al. (2015) also found no significant differences in the number and the area percentage of microglia, when comparing young and old adults (18-month-old rats) (Tan et al., 2015), which is consistent with our results. The controversies found in the number of cells during the aging process are also shown in studies on retinal ganglion cells (RGCs). ...
Chapter
Glaucoma is a neurodegenerative disorder characterized by the loss of retinal ganglion cells and optic nerve fibers, resulting in the loss of visual field. Primary open-angle glaucoma (POAG) is the most prevalent subtype of glaucoma. Recent genome-wide association studies (GWASs) identified more than 100 variants associated with POAG and multiple loci associated with endophenotypes including the disc area, vertical cup-to-disc ratio (VCDR), and intraocular pressure (IOP). Especially, several GWASs reported the association between VCDR and variants near CDKN2B/CDKN2B-AS1, ATOH7, and CHEK2, and between IOP and variants near TMCO1, CAV1/CAV2, GAS7, and ARHGEF12. However, the effect of each variant on endophenotypes is modest; therefore, it is useful to construct a genetic risk score (GRS) based on the effect on endophenotypes by combining susceptible genetic variants. Several studies demonstrated that higher GRS was closely associated with endophenotypes including the VCDR, IOP, and age of diagnosis. Henceforth, by quantifying GRS, identification of high risk group before the disease onset, prediction of visual prognosis and early intervention may be possible.
... These variations could be attributed to differences in species, age, and gender and require further investigation to clarify. 3,[26][27][28][29] Moreover, it is prudent to consider the observed IOP threshold in this study when designing AOH models and conducting related research. ...
Article
Full-text available
Purpose: To assess the correlation between intraocular pressure (IOP) levels and retinal ganglion cell (RGC) loss across different fixed-duration episodes of acute ocular hypertension (AOH). Methods: AOH was induced in Thy1-YFP-H transgenic mice by inserting a needle connected to a saline solution container into the anterior chamber. Thirty-one groups were tested, each comprising three to five mice exposed to IOP levels ranging from 50 to 110 mm Hg in 5/10 mm Hg increments for 60/90/120 minutes and a sham control group. The YFP-expressing RGCs were quantified by confocal scanning laser ophthalmoscopy, whereas peripapillary ganglion cell complex thickness was measured using spectral-domain optical coherence tomography. Changes in RGC count and GCCT were determined from values measured 30 days after AOH relative to baseline (before AOH). Results: In the 60-minute AOH groups, RGC loss varied even when IOP was increased up to 110 mm Hg (36.8%-68.2%). However, for longer durations (90 and 120 minutes), a narrow range of IOP levels (60-70 mm Hg for 90-minute duration; 55-65 mm Hg for 120-minute duration) produced a significant difference in RGC loss, ranging from <25% to >90%. Additionally, loss of YFP-expressing RGCs was comparable to that of total RGCs in the same retinas. Conclusions: Reproducible RGC loss during AOH depends on precise durations and IOP thresholds. In the current study, the optimal choice is an AOH protocol set at 70 mm Hg for a duration of 90 minutes. Translational relevance: This study can assist in determining the optimal duration and intensity of IOP for the effective utilization of AOH models.
... Acute increases in IOP lead to nerve fiber layer thinning with a decrease in RGC soma number in rats (14,52,55) and mice (16), and degeneration of the optic nerve (18). Interestingly, there have also been reports of retinal nerve fiber layer thickening that is resolved by 3 weeks in rats exposed to 8 hours of IOP at 50 mmHg (56). ...
Article
Full-text available
The maintenance of intraocular pressure (IOP) is critical to preserving the pristine optics required for vision. Disturbances in IOP can directly impact the optic nerve and retina, and inner retinal injury can occur following acute and chronic IOP elevation. There are a variety of animal models that have been developed to study the effects of acute and chronic elevation of IOP on the retina, retinal ganglion cell (RGC) morphology, intracellular signaling, gene expression changes, and survival. Acute IOP models induce injury that allows for the study of RGC response to well characterized injury and potential recovery. This review will focus on the initial impact of acute IOP elevation on RGC injury and recovery as these early responses may be the best targets for potential therapeutic interventions to promote RGC survival in glaucoma.
... [24][25][26] Our previous findings showed that the activation of microglial cells emerged at the early stage of ischemia/reperfusion in rat retinae. 27 In addition, minocycline, an inhibitor of microglia cells, obviously suppressed neurodegeneration in ischemic and glaucomatous models. 28,29 Based on these evidences, we hypothesize an association between microglia activation and the pattern of Act-MMP3 expression, and further postulate this pattern of Act-MMP3 expression as an underlying mechanism behind selective neuronal loss in GCLs in acute RI/R. ...
Article
Full-text available
Purpose—Evidences suggest that during ischemia/reperfusion events, neuronal loss in ganglion cell layers (GCLs) occurs initially in the peripheral retinae followed by the central. However, which key molecule or factor mediates this selective loss needs elucidation. In the present study, we detected the regional expression of active matrix metalloproteinase 3 (Act-MMP3) in the central and peripheral rat retinae following acute retinal ischemia/reperfusion (RI/R) injury and explored the effects and mechanisms of this regional expression on the selective neuronal loss in GCLs. Methods—QPCR and Western Blotting were used to detect the expression of Act-MMP3 in the central part and peripheral part of the adult rat retinae. Immunofluorescence and double immunofluorescence were used to assess the number of NeuN-positive cells in the GCLs and Iba-1+CD 68-positive cells were determined. Additionally, the Linear-regression analysis was performed to test the correlation between the ODV of Act-MMP3 and the neuronal loss in the GCLs/Iba-1+CD 68 positive cells in retinae. Results—An evident up-regulation of active matrix metalloproteinase 3 (Act-MMP3) in peripheral retinae preceded to that in central region following acute RI/R. We found Act-MMP3 up-regulation to be associated with the selective neuronal loss in GCLs (central:r=0.7566, p<0.0001, r²=0.5724; peripheral:r=0.8241, p<0.0001, r2=0.6792). Suppressing Act-MMP3 ameliorated the selective neuronal loss in GCLs following acute RI/R. Furthermore, the activation of microglia in the peripheral retinae also preceded to that in the central and was found to be correlated with the regional expression of Act-MMP3 (Central: r=0.8540, p<0.0001, r²=0.7294; Peripheral: r=0.7820, p<0.0001, r²=0.6116.). Suppressing Act-MMP3 ameliorated the microglia regional activation following acute RI/R. Conclusion—The regional expression of Act-MMP3 in the rat retinae may contribute to the selective neuronal loss in GCLs and microglia regional activation in acute RI/R.
Article
Full-text available
Glaucoma is a neurodegenerative disease of the retina characterized by the irreversible loss of retinal ganglion cells (RGCs) leading to visual loss. Degeneration of RGCs and loss of their axons, as well as damage and remodeling of the lamina cribrosa are the main events in the pathogenesis of glaucoma. Different molecular pathways are involved in RGC death, which are triggered and exacerbated as a consequence of a number of risk factors such as elevated intraocular pressure (IOP), age, ocular biomechanics, or low ocular perfusion pressure. Increased IOP is one of the most important risk factors associated with this pathology and the only one for which treatment is currently available, nevertheless, on many cases the progression of the disease continues, despite IOP control. Thus, the IOP elevation is not the only trigger of glaucomatous damage, showing the evidence that other factors can induce RGCs death in this pathology, would be involved in the advance of glaucomatous neurodegeneration. The underlying mechanisms driving the neurodegenerative process in glaucoma include ischemia/hypoxia, mitochondrial dysfunction, oxidative stress and neuroinflammation. In glaucoma, like as other neurodegenerative disorders, the immune system is involved and immunoregulation is conducted mainly by glial cells, microglia, astrocytes, and Müller cells. The increase in IOP produces the activation of glial cells in the retinal tissue. Chronic activation of glial cells in glaucoma may provoke a proinflammatory state at the retinal level inducing blood retinal barrier disruption and RGCs death. The modulation of the immune response in glaucoma as well as the activation of glial cells constitute an interesting new approach in the treatment of glaucoma.
Article
Age and intraocular pressure (IOP) are the two most important risk factors for the development and progression of open-angle glaucoma. While IOP is commonly considered in models of experimental glaucoma (EG), most studies use juvenile or adult animals and seldom older animals which are representative of the human disease. This paper provides a concise review of how retinal ganglion cell (RGC) loss, the hallmark of glaucoma, can be evaluated in EG with a special emphasis on serial in vivo imaging, a parallel approach used in clinical practice. It appraises the suitability of EG models for the purpose of in vivo imaging and argues for the use of models that provide a sustained elevation of IOP, without compromise of the ocular media. In a study with parallel cohorts of adult (3-month-old, equivalent to 20 human years) and old (2-year-old, equivalent to 70 human years) mice, we compare the effects of elevated IOP on serial ganglion cell complex thickness and individual RGC dendritic morphology changes obtained in vivo. We also evaluate how age modulates the impact of elevated IOP on RGC somal and axonal density in histological analysis as well the density of melanopsin RGCs. We discuss the challenges of using old animals and emphasize the potential of single RGC imaging for understanding the pathobiology of RGC loss and evaluating new therapeutic avenues.
Article
Glaucoma is the second leading cause of blindness worldwide. This multifactorial, neurodegenerative group of diseases is characterized by the progressive loss of retinal ganglion cells (RGCs) and their axons, leading to irreversible visual impairment and blindness. There is a huge unmet and urging need for the development of new and translatable strategies and treatment options to prevent this progressive loss of RGC. Accumulating evidence points towards a critical role of neuroinflammation, in particular microglial cells, in the pathogenesis of glaucoma. Leukotrienes are mediators of neuroinflammation and are involved in many neurodegenerative diseases. Therefore, we tested the leukotriene receptors CysLT1R/GPR17-selective antagonist Montelukast (MTK) for its efficacy to modulate the reactive state of microglia in order to ameliorate RGCs loss in experimental glaucoma. Ocular hypertension (OHT) was induced unilaterally by injection of 8 μm magnetic microbead (MB) into the anterior chamber of female Brown Norway rats. The contralateral, untreated eye served as control. Successful induction of OHT was verified by daily IOP measurement using a TonoLab rebound tonometer. Simultaneously to OHT induction, one group received daily MTK treatment and the control group vehicle solution by oral gavage. Animals were sacrificed 13–15 days after MB injection. Retina and optic nerves (ON) of OHT and contralateral eyes were analyzed by immunohistochemistry with specific markers for RGCs (Brn3a), microglial cells/macrophages (Iba1 and CD68), and cysteinyl leukotriene pathway receptors (CysLT1R and GPR17). Protein labeling was documented by confocal microscopy and analyzed with ImageJ plugins. Further, mRNA expression of genes of the inflammatory and leukotriene pathway was analyzed in retinal tissue. MTK treatment resulted in a short-term IOP reduction at day 2, which dissipated by day 5 of OHT induction in MTK treated animals. Furthermore, MTK treatment resulted in a decreased activation of Iba1⁺ microglial cells in the retina and ON, and in a significantly increased RGC survival in OHT eyes. Within the retina, GPR17 and CysLT1R expression was demonstrated in single RCGs and in microglial cells respectively. Further, increased mRNA expression of pro-inflammatory genes was detected in OHT induced retinas. In the ON, OHT induction increased the number of GPR17⁺ cells, showing a significant reduction following MTK treatment. This study shows for the first time a significantly increased RGC survival in an acute OHT model following treatment with the leukotriene receptor antagonist MTK. These results strongly suggest a neuroprotective effect of MTK and a potential new therapeutic strategy for glaucoma treatment.
Chapter
Glaucoma is an age-related neurodegenerative disease that begins at the onset of aging. In this disease, there is an involvement of the immune system and therefore of the microglia. The purpose of this study is to evaluate the microglial activation using a mouse model of ocular hypertension (OHT) at the onset of aging. For this purpose, we used both naive and ocular hypertensives of 15-month-old mice (early stage of aging). In the latter, we analyzed the OHT eyes and the eyes contralateral to them to compare them with their aged controls. In the eyes of aged naive, aged OHT and aged contralateral eyes, microglial changes were observed compared to the young mice, including: (i) aged naive vs young naive: An increased soma size and vertical processes; (ii) aged OHT eyes vs young OHT eyes: A decrease in the area of the retina occupied by Iba-1 cells and in vertical processes; and (iii) aged contralateral vs young contralateral: A decrease in the soma size and arbor area and an increase in the number of microglia in the outer segment layer. Aged OHT eyes and the eyes contralateral to them showed an up-regulation of the CD68 expression in the branched microglia and a down-regulation in the MHCII and P2RY12 expression with respect to the eyes of young OHT mice. Conclusion: in the early phase of aging, morphological microglial changes along with changes in the expression of MHCII, CD68 and P2RY12, in both naive and OHT mice. These changes appear in aged OHT eyes and the eyes contralateral to them eyes.
Article
Full-text available
Retinal ganglion cell apoptosis and optic nerve degeneration are prevalent in aged patients, which may be related to the decrease in bone marrow (BM) stem cell number/function because of the possible cross‐talk between the two organs. This pathological process is accelerated by retinal ischaemia‐reperfusion (I/R) injury. This study investigated whether young BM stem cells can regenerate and repair the aged retina after acute I/R injury. Young BM stem cell antigen 1 positive (Sca‐1⁺) or Sca‐1⁻ cells were transplanted into lethally irradiated aged recipient mice to generate Sca‐1⁺ and Sca‐1⁻ chimaeras, respectively. The animals were housed for 3 months to allow the young Sca‐1 cells to repopulate in the BM of aged mice. Retinal I/R was then induced by elevation of intraocular pressure. Better preservation of visual function was found in Sca‐1⁺ than Sca‐1⁻ chimaeras 7 days after injury. More Sca‐1⁺ cells homed to the retina than Sca‐1⁻ cells and more cells differentiated into glial and microglial cells in the Sca‐1⁺ chimaeras. After injury, Sca‐1⁺ cells in the retina reduced host cellular apoptosis, which was associated with higher expression of fibroblast growth factor 2 (FGF2) in the Sca‐1⁺ chimaeras. Young Sca‐1⁺ cells repopulated the stem cells in the aged retina and diminished cellular apoptosis after acute I/R injury through FGF2 and Akt signalling pathways.
Article
Full-text available
To study changes in scleral structure induced by chronic experimental intraocular pressure elevation in mice. We studied the effect of chronic bead-induced glaucoma on scleral thickness, collagen lamellar structure, and collagen fibril diameter distribution in C57BL/6 (B6) and CD1 mice, and in collagen 8α2 mutant mice (Aca23) and their wild-type littermates (Aca23-WT) using electron and confocal microscopy. In unfixed tissue, the control B6 peripapillary sclera was thicker than in CD1 mice (p<0.001). After 6 weeks of glaucoma, the unfixed CD1 and B6 sclera thinned by 9% and 12%, respectively (p<0.001). The fixed sclera, measured by electron microscopy, was significantly thicker in control Aca23 than in B6 or CD1 mice (p<0.05). The difference between fresh and fixed scleral thickness was nearly 68% in untreated control B6 and CD1 mice, but differed by only 10% or less in fresh/fixed glaucoma scleral comparisons. There were 39.3±9.6 lamellae (mean, standard deviation) in control sclera, categorized as 41% cross-section, 24% cellular, 20% oblique, and 15% longitudinal. After glaucoma, mean peripapillary thickness significantly increased in fixed specimens of all mouse strains by 10.3 ±4.8 µm (p=0.001) and the total number of lamellae increased by 18% (p=0.01). The number of cellular and cross-section lamellae increased in glaucoma eyes. After glaucoma, there were more small and fewer large collagen fibrils (p<0.0001). Second harmonic generation imaging showed that the normal circumferential pattern of collagen fibrils in the peripapillary sclera was altered in significantly damaged glaucomatous eyes. Dynamic responses of the sclera to experimental mouse glaucoma may be more important than baseline anatomic features in explaining susceptibility to damage. These include decreases in nonfibrillar elements, alterations in lamellar orientation, an increased number of smaller collagen fibrils and fewer larger fibrils, and relative increase in the number of scleral fibroblast layers.
Article
Full-text available
Primary open angle glaucoma (POAG) is a neurodegenerative disease characterized by physiological intraocular hypertension that causes damage to the retinal ganglion cells (RGCs). In the past, RGC damage in POAG was suggested to have been attributed to RGC apoptosis. However, in the present study, we applied a model closer to human POAG through the use of a chronic hypertensive glaucoma model in rhesus monkeys to investigate whether another mode of progressive cell death, autophagy, was activated in the glaucomatous retinas. First, in the glaucomatous retinas, the levels of LC3B-II, LC3B-II/LC3B-I and Beclin 1 increased as demonstrated by Western blot analyses, whereas early or initial autophagic vacuoles (AVi) and late or degraded autophagic vacuoles (AVd) accumulated in the ganglion cell layer (GCL) and in the inner plexiform layer (IPL) as determined by transmission electron microscopy (TEM) analysis. Second, lysosome activity and autophagosome-lysosomal fusion increased in the RGCs of the glaucomatous retinas, as demonstrated by Western blotting against lysosome associated membrane protein-1 (LAMP1) and double labeling against LC3B and LAMP1. Third, apoptosis was activated in the glaucomatous eyes with increased levels of caspase-3 and cleaved caspase-3 and an increased number of TUNEL-positive RGCs. Our results suggested that autophagy was activated in RGCs in the chronic hypertensive glaucoma model of rhesus monkeys and that autophagy may have potential as a new target for intervention in glaucoma treatment.
Article
Full-text available
Multiple sclerosis (MS) is often accompanied by optic nerve inflammationAndsome patients experience permanent vision loss. We examined if the grade of optic nerve infiltration and demyelination affects the severity of clinical signs in an experimental autoimmune encephalomyelitis (EAE) model. The loss of retinal ganglion cells (RGC) and alterations in glia activity were also investigated. C57BL/6 mice were immunized with peptide MOG35-55in complete Freund's adjuvant (CFA) and controls received PBS in CFA. Then 23 days post immunization eyes were prepared for flatmounts and stained with Nissl to evaluated neuronal density. Clinical EAE symptoms as well as cell infiltration and demyelination in the optic nerve were examined. Retinal sections were stained with hematoxylin and eosin and silver stain. Immunohistochemistry was usedto label RGCs (Brn-3a), apoptotic cells (caspase 3), macroglia (glial fibrillary acidic protein (GFAP)), microglia (Iba1), macrophages (F 4/80) and interleukin-6 (IL-6) secretion. EAE symptoms started at day 8 and peaked at day 15. Cell infiltrations (P = 0.0047) and demyelination (P = 0.0018) of EAE nerves correlated with the clinical score (r> 0.8). EAE led to a significant loss of RGCs (P< 0.0001). Significantly more caspase 3+ cells were noted in these animals (P = 0.0222). They showed an increased expression of GFAP (P< 0.0002) and a higher number of microglial cells (P< 0.0001). Also more macrophages and IL-6 secretion were observed in EAE mice. MOG immunization leads to optic neuritis and RGC loss. EAE severity is related to the severity of optic nerve inflammation and demyelination. EAE not only affects activation of apoptotic signals, but also causes a glial response in the retina.
Article
Full-text available
Purpose: We examined the signaling mechanisms involved in δ-opioid-receptor agonist, SNC-121-mediated attenuation of TNF-α-induced matrix metalloproteinase-2 (MMP-2) secretion from human optic nerve head (ONH) astrocytes. Methods: Human ONH astrocytes were treated with SNC-121 (1 μmol/L) for 15 minutes followed by TNF-α (25 ng/mL) treatment for 6 or 24 hours. Cells were pretreated with inhibitors of p38 mitogen-activated protein (MAP) kinase (SB-203580) or NF-κB (Helenalin) prior to TNF-α treatment. Changes in phosphorylation and expression of p38 MAP kinase, IκBα, NF-κB, and MMP-2 were measured by Western blotting. Translocation of NF-κB was determined by immunocytochemistry. Results: TNF-α treatment increased MMP-2 secretion from ONH astrocytes to 236% ± 17% and 142% ± 8% at 6 and 24 hours, respectively; while SNC-121 treatment reduced MMP-2 secretion to 149% ± 11% and 108% ± 7% at 6 and 24 hours, respectively. The SNC-121-mediated inhibitory response was blocked by the δ-opioid-receptor antagonist naltrindole. TNF-α treatment resulted in a sustained phosphorylation of p38 MAP kinase up to 24 hours (226% ± 15% over control levels), which was reduced to 150% ± 20% by SNC-121 treatment. TNF-α treatment increased the expression of NF-κB to 179% ± 21% and 139% ± 6% at 6 and 24 hours, respectively, which was significantly blocked by SNC-121 treatment. Furthermore, TNF-α-induced MMP-2 secretion was blocked by 100% and 78% in the presence of SB-203580 and Helenalin, respectively. Conclusions: Evidence is provided that SNC-121 attenuated TNF-α-induced MMP-2 secretion from ONH astrocytes. Data also supported the idea that p38 MAP kinase and NF-κB played central roles in TNF-α-induced MMP-2 secretion, and both were negatively regulated by SNC-121.
Article
Full-text available
Glaucoma, frequently associated with elevated intraocular pressure (IOP), is one of the leading causes of blindness. We sought to establish a mouse model of ocular hypertension to mimic human high-tension glaucoma. Here laser illumination is applied to the corneal limbus to photocoagulate the aqueous outflow, inducing angle closure. The changes of IOP are monitored using a rebound tonometer before and after the laser treatment. An optomotor behavioral test is used to measure corresponding changes in visual capacity. The representative result from one mouse which developed sustained IOP elevation after laser illumination is shown. A decreased visual acuity and contrast sensitivity is observed in this ocular hypertensive mouse. Together, our study introduces a valuable model system to investigate neuronal degeneration and the underlying molecular mechanisms in glaucomatous mice.
Article
Full-text available
Glaucoma is conventionally defined as a chronic optic neuropathy characterized by progressive loss of retinal ganglion cells (RGCs) and optic nerve fibers. Although glaucoma is often associated with elevated intraocular pressure (IOP), significant IOP reduction does not prevent progression of the disease in some glaucoma patients. Thus, exploring IOP-independent mechanisms of RGC loss is important. We describe chronic systemic administration of aldosterone and evaluate its effect on RGCs in rat. Aldosterone was administered via an osmotic minipump that was implanted subcutaneously into the mid-scapular region. Although systemic administration of aldosterone caused RGC loss associated with thinning of the retinal nerve fiber layer without elevated IOP, the other cell layers appeared to be unaffected. After chronic administration of aldosterone, RGC loss was observed at 2 weeks in the peripheral retina and at 4 weeks in the central retina. However, administration of mineralocorticoid receptor blocker prevented RGC loss. These results demonstrate aldosterone is a critical mediator of RGC loss that is independent of IOP. We believe this rat normal-tension glaucoma (NTG) animal model not only offers a powerful system for investigating the mechanism of neurodegeneration in NTG, but can also be used to develop therapies directed at IOP-independent mechanisms of RGC loss.
Article
We have determined whether various risk factors relevant to neural injury in the human central nervous system (CNS) can be modeled in the rat. In a model for CNS injury, we have quantitated the survival of retinal ganglion cells (RGCs) following retinal ischemia/reperfusion. Aging was studied by comparing 2‐year‐old rats with 2‐month‐old rats. Caloric restriction was created by providing food to both young and old animals three days per week for 3 months; controls had continuous access to food. Diabetes was induced by intravenous injection of streptozotocin; controls were injected with citrate buffer. Rats with chronic, moderately elevated intraocular pressure (IOP) were compared with rats with normal IOP. Albino rats and pigmented rats were compared. In all animals, ischemia/reperfusion was produced unilaterally by elevating IOP above systolic pressure for 75 minutes by using anterior chamber cannulation. Loss of RGCs was determined by retrograde labeling with fluorogold. The number of RGCs decreases with age. The remaining RGCs in old rats in both central and peripheral retina were significantly more susceptible to ischemia/reperfusion compared with young rats. Caloric restriction significantly protected against loss of RGCs in the peripheral retina in both young and old rats. In diabetic rats, RGCs throughout the retina were more susceptible to ischemia/reperfusion compared with control rats. The remaining RGCs in rats with glaucoma (preexisting chronic, moderately elevated IOP) were more susceptible to ischemia/reperfusion damage. Comparing pigmented and albino rats, the loss of RGCs following ischemia/reperfusion did not differ. Our results suggest that aging, diabetes, and glaucoma are risk factors for the loss of RGCs following ischemic damage of the retina and can be modeled in rats. Furthermore, reducing caloric intake appears to be neuroprotective for ischemic damage of the retina at all ages. The underlying cellular and molecular factors that are responsible for these risk factors can be studied by using these expanded models of neural injury.
Article
purpose. To develop a modified adenoassociated viral (AAV) vector capable of efficient transfection of retinal ganglion cells (RGCs) and to test the hypothesis that use of this vector to express brain-derived neurotrophic factor (BDNF) could be protective in experimental glaucoma. methods. Ninety-three rats received one unilateral, intravitreal injection of either normal saline (n = 30), AAV-BDNF-woodchuck hepatitis posttranscriptional regulatory element (WPRE; n = 30), or AAV-green fluorescent protein (GFP)-WPRE (n = 33). Two weeks later, experimental glaucoma was induced in the injected eye by laser application to the trabecular meshwork. Survival of RGCs was estimated by counting axons in optic nerve cross sections after 4 weeks of glaucoma. Transgene expression was assessed by immunohistochemistry, Western blot analysis, and direct visualization of GFP. results. The density of GFP-positive cells in retinal wholemounts was 1,828 ± 299 cells/mm² (72,273 ± 11,814 cells/retina). Exposure to elevated intraocular pressure was similar in all groups. Four weeks after initial laser treatment, axon loss was 52.3% ± 27.1% in the saline-treated group (n = 25) and 52.3% ± 24.2% in the AAV-GFP-WPRE group (n = 30), but only 32.3% ± 23.0% in the AAV-BDNF-WPRE group (n = 27). Survival in AAV-BDNF-WPRE animals increased markedly and the difference was significant compared with those receiving either AAV-GFP-WPRE (P = 0.002, t-test) or saline (P = 0.006, t-test). conclusions. Overexpression of the BDNF gene protects RGC as estimated by axon counts in a rat glaucoma model, further supporting the potential feasibility of neurotrophic therapy as a complement to the lowering of IOP in the treatment of glaucoma.