- Wiley Online Library

advertisement
Ocular perfusion and age-related
macular degeneration
Thomas A. Ciulla, Alon Harris and Bruce J. Martin
Indiana University School of Medicine, Indianapolis, Indiana, USA
ABSTRACT.
Purpose: To review the role of ocular perfusion in the pathophysiology of agerelated macular degeneration (AMD), the leading cause of irreversible blindness
in the industrialized world.
Methods: Medline search of the literature published in English or with English
abstracts from 1966 to 2000 was performed using various combinations of relevant key words.
Results: Vascular defects have been identified in both nonexudative and exudative AMD patients using fluorescein angiographic methods, laser Doppler
flowmetry, indocyanine green angiography, and color Doppler imaging.
Conclusion: Although these studies lend some support to the vascular pathogenesis of AMD, it is not possible to determine if the choroidal perfusion abnormalities play a causative role in nonexudative AMD, if they are simply an association with another primary alteration, such as a primary RPE defect or a
genetic defect at the photoreceptor level, or if they are more strongly associated
with one particular form of this heterogeneous disease. Further study is warranted.
Key words: age-related macular degeneration – ocular perfusion – blood flow – choriod – choriocapiIlaris – retinal circulation.
Acta Ophthalmol. Scand. 2001: 79: 108–115
Copyright c Acta Ophthalmol Scand 2001. ISSN 1395-3907
A
ge related macular degeneration
(AMD) is the leading cause of irreversible visual loss in the United States,
occurring in over 10% of the population
aged 65 to 74 years and over 25% of the
population over the age of 74 years (Leibowitz et al. 1980). There are two types of
macular degeneration. The nonexudative
form involves atrophic and hypertrophic
changes in the retinal pigment epithelium
(RPE) underlying the central retina or
macula, drusen. Patients with nonexudative AMD can progress to the exudative
form of AMD, in which choroidal neovascular membranes (CNVM) develop
under the retina, leak fluid and blood,
and ultimately cause a blinding ‘‘disciform’’ scar in and under the retina. Nonexudative AMD occurs in approximately
27% of patients over 75 years, and exudative AMD occurs in nearly 5% of this
group (Klein et al. 1992). Overall, ap-
108
proximately 10–20% of patients with
nonexudative AMD progress to the exudative form, which is responsible for
most of the estimated 1.2 million cases of
severe visual loss from AMD (Hyman et
al. 1993; Tielsch et al. 1995). Also, it
should be noted that AMD is a bilateral
disorder; CNVM develop in over onefourth (26%) of fellow eyes that are initially free of exudative AMD over a fiveyear period (MPS 1993). As the population in the United States ages, visual loss
from AMD will become even more prevalent.
Nonexudative AMD is much more
common than exudative AMD and is
usually a precursor of exudative AMD.
The nonexudative form involves a variety
of presentations including hard drusen,
soft drusen and geographic (areolar)
atrophy of the RPE. Hard drusen are associated with localized dysfunction of the
RPE, while soft drusen are associated
with diffuse dysfunction of the RPE
(Bressler et al. 1988). Although more typically associated with exudative AMD, severe visual loss can occur from nonexudative AMD, particularly when geographic atrophy of the RPE develops in
the fovea to cause a central scotoma (Ciulla et al. 1998). Unfortunately, there is
no widely accepted treatment modality
for this devastating form of AMD, although some investigators have suggested
a protective effect from various micronutrients and vitamins and other investigators are studying laser treatment
of drusen (Ciulla et al. 1998).
Although the exudative form is treatable, treatment efficacy is currently low.
Currently, the only well-studied and
widely-accepted method of treatment is
laser photocoagulation of the CNVM as
demonstrated in several well designed
clinical trials, including the Macular
Photocoagulation Study (MPS), a group
of multicenter, randomized, controlled
clinical trials of laser photocoagulation
of choroidal neovascularization in patients with AMD, the presumed ocular
histoplasmosis syndrome, or in idiopathic
cases. The MPS showed that photocoagulation effectively prevented large decreases in visual acuity compared to observation. However, only 13–26% of patients with exudative AMD show welldemarcated ‘‘classic’’ CNVM (recognized
on fluorescein angiography as discreet
early hyperfluorescence with late leakage)
amenable to laser treatment and at least
half of these patients suffer from persistent or recurrent CNVM formation within
two years. Patients with poorly demarcated (due to subretinal hemorrhage, for
example) or ‘‘occult’’ CNVM (recognized
on fluorescein angiography as a fibrovascular pigment epithelial detachment
or late leakage of undetermined source)
make up the majority of patients with exudative AMD and are not eligible for
Fig. 1. According to the traditonal theory, primary genetic, dietary, or
photo-oxidative stress leads to RPE dysfunction, which consequently
leads to choriocapillaris atrophy and choroidal perfusion defects.
laser photocoagulation (Freund et al.
1993; Moisseiev et al. 1995; MPS 1982;
MPS 1986; MPS 1991).
Pathogenesis of AMD
Traditional theory
Several theories of pathogenesis have
been proposed and these include primary
RPE and Bruch’s membrane senescence,
primary genetic defects, and primary
ocular perfusion abnormalities. Oxidative insults have also been proposed as a
contributing factor. Traditionally, investigators have felt that senescence of the
RPE, which metabolically supports and
maintains the photoreceptors, leads to
AMD (Eagle 1984; Young 1987). It has
been felt that the senescent RPE accumulates metabolic debris as remnants of incomplete degradation from phagocytosed
rod and cone membranes and that progressive engorgement of these RPE cells
leads to drusen formation with subsequent progressive further dysfunction
of the remaining RPE (Eagle 1984;
Young 1987). (Figs. 1 & 2).
Bruch’s membrane, thickened with
drusen, could be predisposed to crack
formation (Green et al. 1985; Sarks
1976). Calcification and fragmentation of
Bruch’s membrane is more prominent in
eyes with exudative AMD, and it is
thought that these defects in Bruch’s
membrane could facilitate development
of CNVM (Spraul & Grossniklaus 1997).
This theory is supported by findings in
Fig. 2. According to the traditional theory, senescent RPE is susceptible
to genetic, dietary, or photo-oxidative stress, which in turn leads to
failure to metabolize photoreceptor outer segments that are constantly
being shed. This leads to drusen formation. In addition, RPE dysfunction or atrophy leads to choriocapillaris atrophy.
myopic degeneration and angioid streaks
in which CNVM develop through breaks
in Bruch’s membrane. The well-known
primate-laser model, developed by Ryan,
may also support this theory for CNVM
(Ishibashi et al. 1995, 1985, 1987; Miller
et al. 1990; Nishimura et al. 1990; Ohkuma & Ryan 1982, 1983; Ryan 1979,
1982). In this model, high intensity laser
burns are used to create ruptures in
Bruch’s membrane/RPE complex to initiate a repair process in the fundus that
results in the development of subretinal
neovascularization (Miller et al. l990).
The exact stimulus for CNVM formation
is unclear; it is possible that macrophages
involved in the initial response to Bruch’s
membrane injury secrete angiogenic
growth factors (Ishibashi et al. 1985; Nishimura et al. 1990).
Another potential mechanism by
which CNVM could develop in response
to fragmentation of Bruch’s membrane
could relate to matrix metalloproteinases
(MMP) which are extracellular matrix
degrading enzymes that may play a key
role in angiogenesis and CNVM formation (Steen et al. 1998). Bruch’s membrane has been shown to contain tissue
inhibitor of metalloproteinases (TlMP)-3
(Faris et al. 1997) and eyes with AMD
have been shown to have abnormal levels
of TIMP-3 compared to normal agematched control eyes (Kamei & Holleyfield 1999); calcification and fragmentation observed in Bruch’s membrane
may represent a breach in this antiangiogenic barrier, facilitating CNVM devel-
opment. Whatever the initial stimulus for
CNVM formation, it is clear that angiogenic growth factors are ultimately involved. Surgically excised and post mortem CNVM tissue, as well RPE cells, have
been shown to be immunoreactive for
various growth factors thought to be
angiogenic, including vascular endothelial growth factor (VEGF), transforming growth factor-beta (TGF-b),
platelet derived growth factor (PDGF)
and basic fibroblast growth factor (FGF)
(Amin et al. 1994: Kvanta 1995; Lopez et
al. 1996; Reddy et al. 1995).
Genetic Defects
Yet another theory involves genetic defects. A variety of genes have been suggested. For example, some investigators
recently reported that 16% of AMD patients in their study had a genetic defect
in a gene encoding a retinal rod protein,
the ABCR gene, which has also been
found to be defective in Stargardt’s disease (Allikmets et al. 1997). However,
there have been two more recent publications suggesting that the ABCR mutations might not be linked to AMD (De
La Paz et al. 1999; Stone et al. 1998).
There have also been recent reports of a
genetic association between AMD and
apolipoprotein E, a protein that plays a
role in central nervous system lipid
homeostasis (Klaver et al. 1998; Souied et
al. 1998). Investigators are also studying
other hereditary dystrophies with some
109
features similar to AMD, such as Malattia Leventinese and Doyne’s Honeycomb
retinal dystrophy, which are characterized
by drusen and have been associated with
a single mutation in the gene for EGFcontaining fibrillin-like extracellular matrix protein 1 (Stone et al. 1999). Sorsby’s
dystrophy, an autosomal dominant disorder that presents in the fifth decade,
shows several similarities to AMD, including submacular exudates and hemorrhages initially followed by cicatrization,
and a point mutation on the TIMP-3
gene has been reported (Weber et al.
1994). Another group, however, found no
evidence of linkage or association between the TIMP-3 gene in 38 multiplex
families with AMD (De La Paz et al.
1997).
group of investigators showed correlation
between the regions of sparring in annular maculopathy and the spatial distribution of macular pigments (Weiter et al.
1988). Furthermore, factors known to
decrease macular pigment optical density
(MPOD) levels, such as cigarette smoking (Hammond et al. 1996), light iris
color (Ciulla et al. 2000; Hammond et al.
1996), and female gender (Hammond et
al. 1996), have also been implicated to increase the risk of AMD in epidemiologic
studies; this parallel is directional, with
factors that decrease MPOD leading to
increased risk of AMD and factors that
increase MPOD leading to decreased risk
of AMD, consistent with a potential protective role of macular pigments in AMD.
Oxidative Insults
Ocular Perfusion Defects
in AMD
Oxidative insults have also been proposed
as a contributing factor and this may involve the macular pigments, lutein and
zeaxanthin, which are primarily obtained
from dark green, leafy vegetables and account for the yellow pigmentation of the
macula lutea (Bone et al. 1993, 1985).
Macular pigment is of dietary origin as
humans and non-human primates do not
synthesize carotenoids de novo (Malinow
et al. 1980; Neuringer et al. 1999). Macular pigment has been hypothesized to
play a protective role against the development of AMD through the limitation of
oxidative insults (Katz et al. 1982;
Schalch 1992; Seddon & Hennekens
1994; Snodderly et al. 1984) by filtering
out harmful wavelengths of light (Bone et
al. 1985) or by the antioxidant properties
of its components (Katz et al. 1982;
Schalch 1999).
A recent study showed that primates
raised on carotenoid-depleted diets had a
significantly increased incidence of angiographic transmission defects in the macular regions (Neuringer et al. 1999). Previous studies have shown that a higher
dietary intake of L and Z has been associated with a lower risk for AMD (Seddon
et al. 1994; West et al. 1994). Another
link between macular pigment and AMD
derives from observations of atrophic
AMD. The central fovea, which contains
the highest concentrations of macular
pigment, is often spared from atrophy
until late in progression, causing some investigators to postulate that macular pigments account for the relative resistance
of this area to degenerative change; one
110
With regard to the role of choroidal perfusion defects, it is well known that the
choriocapillaris supplies the metabolic
needs of the retinal pigment epithelium
and the outer retina; a primary perfusion
defect in the choriocapillaris could account for some of the physiologic and
pathologic changes in AMD. For example, both acute ischemia and the subsequent reperfusion of the brain are associated with CNS cell death (Chen et al.
1997; Gillardon et al. 1996; Leib et al.
1996; Macaya 1996; Nickells 1996; Quigley et al. 1995). Cell death after ischemia
occurs primarily by apoptosis, especially
when the insult is mild (Chen et al. 1997;
Gillardon et al. 1996; Leib et al. 1996;
Macaya 1996). Mild ischemia is postulated to provoke precisely such a cellular
event in retinal ganglion cells in glaucoma (Chen et al. 1997; Gillardon et al.
1996; Harris et al. 2000; Leib et al. 1996;
Macaya 1996; Nickells 1996; Quigley et
al. 1995), which is characterized by slow
loss of these cells over years, and some
forms of AMD (such as geographic atrophy of the RPE characterized by progressive loss of RPE cells) could have a
similar pathogenesis to glaucoma.
In the vascular model for AMD pathogenesis proposed by Friedman, it is
theorized that lipid deposition in sclera
and Bruch’s membrane leads to scleral
stiffening and impaired choroidal perfusion, which would in turn adversely affect metabolic transport function of the
retinal pigment epithelium (Friedman
1997; Friedman et al. 1995). The im-
paired RPE cannot metabolize and transport material shed from the photoreceptors, leading to accumulation of metabolic debris and drusen (Friedman 1997;
Friedman et al. 1995). (Figs. 3–5). This
theory is supported by studies demonstrating an association between increased
scleral rigidity and AMD (Friedman et
al. 1989). In addition, AMD, with its
widely varying clinical presentations,
may actually represent several distinct
disorders that have yet to be more clearly
differentiated on a pathogenic basis; it is
possible that RPE senescence represents
the primary derangement in some subforms, choroidal perfusion defects represent the primary derangement in other
subforms, and perhaps specific genetic
defects represent the primary defect in yet
other subforms. Epidemiological risk
factors, such as tobacco use (Christen et
al. 1996; Seddon et al. 1996), blue light
or sunlight exposure (Cruickshanks et al.
1993) and nutritional factors (MaresPerlman et al. 1995; Seddon et al. 1994)
could represent environmental influences
that exert a detrimental secondary effect
on individuals with any of the underlying
primary derangements noted above. Of
note, many of the risk factors for AMD
are similar to those for cardiovascular
disease, including tobacco use, hypertension, and nutritional factors, lending
further support for a vascular role in
AMD.
The vascular theory is supported by
studies demonstrating delayed choroidal
filling in AMD using conventional angiographic techniques (Boker et al. 1993; Remulla et al. 1995; Zhao et al. 1995). Delayed choroidal filling may have histologic significance as it may correlate with
diffuse thickening of Bruch’s membrane
(Pauleikhoff et al. 1990). This sign also
has functional significance as eyes with
this sign harbor discrete areas of increased threshold on static perimetry
(Chen et al. 1992) and are at risk for loss
of vision (Piguet et al. 1992). Specifically,
in one study, 38% of 32 eyes with this sign
lost two or more lines of vision by two
years compared to 14% of 64 eyes without this sign (Piguet et al. 1992). This difference was related to the greater incidence of geographic atrophy in the patients with delayed choroidal filling;
significantly, the incidence of CNVM was
similar in each group of patients (Piguet
et al. 1992).
It is very difficult to quantify choroidal
blood flow angiographically given the
overlying retinal circulation and multilay-
Fig. 3. According to the vascular theory, primary choroidal perfusion
abnormalities lead to RPE dysfunction and then to AMD.
Fig. 5. Decreased choroidal perfusion impairs RPE transport function
as the RPE must pump against a higher osmotic gradient. The photoreceptor outer segments, which are constantly being shed, are not properly metabolized by the RPE, which leads to drusen formation.
Fig. 4. Choroidal perfusion abnormalities arise as lipid is deposited in
the sclera and Bruch’s membrane, which then leads to collagen and
elastic degeneration, causing scleral stiffening. The increase in scleral
rigidity leads to decreased compliance and increased resistance in the
choroid, which then leads to decreased choroidal perfusion.
Fig. 6. According to the vascular model as proposed by Friedman
(Friedman 1997; Freidman et al. 1995), there is a generalized stiffening
and increase in resistance, not only in the choroidal vasculature, but
also in the cerebral vasculature. If the choroidal resistance increases
more that the cerebral vascular resistance, there is a decrease in choroidal perfusion with an increase in the osmotic gradient against which
the RPE must pump, leading to an accumulation of metabolic debris
in the form of drusen. If the choroidal resistance increases less than the
cereoral vascular resistance, there is higher choroidal perfusion pressure, which facilitates CNVM development.
ered choroidal circulation that complicates analysis. Recently, however, new
technologies have been employed to
corroborate the existence of choroidal
perfusion anomalies in AMD, including
ICG angiography, laser Doppler flowmetry, and color Doppler imaging
(Harris et al. 1999).
One group recently used a new analysis
technique based on indocyanine green
angiography to compare the choroidal
circulation in patients with AMD to a
control group. ICG facilitates study of
the choroidal circulation for several reasons. First, ICG better delineates the
choroidal circulation than fluorescein because the near-infrared light absorbed by
ICG penetrates the retina pigment epithelium better than the shorter wavelength absorbed by fluorescein. Also, unlike fluorescein, ICG is strongly bound to
plasma proteins, which prevents diffusion
of the compound through the fenestrated
choroidal capillaries, and permits better
delineation of choroidal details. This
study showed a statistically significant increased frequency of presumed macular
watershed filling (PMWF), which was described as ‘‘characteristic vertical, angled,
or stellate-shaped zones of early-phase
indocyanine green videoangiographic hypofluorescence, assumed to be hypoperfusion, which disappeared in the early
phase of the angiogram’’ (Ross et al.
l998). The investigators noted that 55.4%
of 74 patients with AMD versus 15.0%
111
of 20 normal control patients exhibited
PMWF. They also note that 59.0% of the
61 patients with AMD-associated choroidal
neovascularization
exhibited
PMWF and that the CNVM arose from
the PMWF zone in 91.7% of these cases.
This analysis approach provides valuable
insight into abnormalities of choroidal
circulation in AMD, although this
method requires subjective assessment for
the presence and location of PMWF.
Another more objective approach has
been employed using a new, area dilution
analysis technique applied to ICG angiography (Ciulla et al. 1998; Harris et al.
1998). Scanning laser ophthalmoscope
angiograms in 21 nonexudative AMD
subjects were compared to 21 agematched control subjects (Ciulla et al.
2000). After correction for eye movements, the digital image analysis system
recorded mean intensity levels at each
area over time. Four areas around the
macula and two areas in the temporal
peripapillary retina were evaluated. Fluorescence density in each block was averaged over time, graphed, and analyzed
for dye appearance time and maximal
fluorescence. In addition, intensity curves
were analyzed by quantifying the slope of
the filling portion of the curves, the
amount of time required to rise 10% and
63% above baseline by graphing intensity
of fluorescence of each area over time. Although the exact concentration of ICG in
each region cannot be determined, simultaneous acquisition of dye dilution curves
from these regions within the choroid facilitates comparison of relative concentrations between these regions. Since the
six analysis regions are identically positioned on each subjects angiogram, the
resulting analysis represents a very objective evaluation of choroidal perfusion
characteristics and does not rely on subjective assessment. This technique showed
statistically significant delayed and heterogeneous filling within the choriocapillaris of nonexudative AMD patients
when compared to normal age-matched
controls, and these changes showed some
macular-region specificity (Ciulla et al.
2000). In addition, the localized choroidal perfusion defects correlated in a
statistically significant fashion with visual
acuity and severity of non-exudative agerelated macular degeneration.
Another group used a new technique
called laser Doppler flowmetry in subjects with nonexudative AMD to show
that the choroidal blood flow was decreased at the center of the fovea com-
112
pared to a control group (Grunwald et al.
1998). This technique cannot be readily
applied outside the foveal center as the
overlying retinal circulation would
Doppler shift the reflected light from the
laser and prevent analysis of the choroid.
Nevertheless, this study and the ICG angiogram studies were consistent in that
this prior study showed alterations in
choroidal flow in the foveal center and
the ICG angiogram studies confirmed
alterations with some macular-region
specificity, although the foveal center itself was not measured.
Color Doppler imaging has been used
to evaluate the retrobulbar vasculature in
AMD; two groups have found statistically significant differences in the central
retinal and posterior ciliary arteries in
patients with AMD compared to controls
(Ciulla et al. 1999; Friedman 1995, 1997).
Critics of the vascular theory point to
literature that suggests that healthy RPE
necessary for maintenance of the choroid
and choriocapillaris (Del Priore et al.
1995, 1996; Henkind & Gartner 1983;
Korte et al. 1984; Nasir et al. 1997; Pollack et al. 1996; Takeuchi et al. 1993) and
that the vascular changes found in AMD
are secondary to primary RPE dysfunction. In primate eyes, in which the RPE
was selectively damaged by intravitreal
ornithine (Takeuchi et al. 1993) and in
porcine eyes in which the RPE was debrided surgically, the choriocapillaris degenerated by two months and one week,
respectively (Del Priore et al. 1995, 1996).
Another group obtained a similar result
in a rabbit model in which the RPE was
damaged by intravenous sodium iodate,
and they also observed absence of the
choriocapillaris histopathologically in
human eyes with retinitis pigmentosa;
they postulated the existence of a diffusable vascular modulation factor, produced by the RPE and responsible for
choriocapillaris fenestrae formation and
maintenance (Henkind & Gartner 1983;
Korte et al. 1984). Some reports suggest
that surgical removal of the RPE in
humans (during removal of choroidal
neovascular membranes) leads to abnormal perfusion of the choriocapillaris, although it is not possible to rule out preexisting atrophy or intraoperative damage (Nasir et al. 1997; Pollack et al.
1996).
Although it may not be possible to definitively determine it the vascular derangements found in AMD are primary
or secondary, findings from color Doppler imaging may support primary vascular
dysfunction. Specifically, it is intriguing
to elicit perfusion deficits in the central
retinal artery of AMD subjects, as one
would not necessarily anticipate a vascular derangement beyond the posterior ciliary arteries, which supply the choroid.
These results suggest that there may be a
more generalized perfusion defect beyond
the choroid in patients with AMD. In addition, the vascular model could account
for development of both the nonexudative and exudative forms of AMD. According to the vascular model, there is a
generalized stiffening and increase in resistance, not only in the choroidal vasculature, but also in the cerebral vasculature (Friedman 1997; Friedman et al.
1995). If the choroidal resistance increases more than the cerebral vascular
resistance, there is a decrease in choroidal
perfusion with an increase in the osmotic
gradient against which the RPE must
pump, leading to an accumulation of
metabolic debris in the form of drusen. If
the choroidal resistance increases less
than the cerebral vascular resistance,
there is higher choroidal perfusion pressure, which facilitates CNVM development. This mechanism could partially account for the development of CNVM in
the presence of Bruch’s membrane senescence or cracks.
Therapies Based on
Blood Flow
It is unclear if improving macular perfusion would lead to improved function
in nonexudative AMD or delayed disease
progression. Friedman originally proposed a clinical study that involves lamellar scleral resection as a means of decreasing scleral rigidity to enhance choroidal perfusion and in turn arrest the
progression of the disease. However, this
proposition generated significant controversy, given the invasive nature of this
procedure and the unsettled role of ocular perfusion defects in the many presentations and types of AMD. Nevertheless,
several currently available topical ophthalmic and systemic pharmaceutical
agents have been shown to enhance ocular blood flow in normal and glaucomatous eyes and these agents could have a
potential role in AMD. In healthy individuals, for example, three different topical beta-blockers, including both Beta1selective and nonselective agents (levobunolol, betaxolol, and timolol) each re-
duced the intraocular pressure (IOP), decreased the retinal arteriovenous passage
time, and increased macular capillary and
epipapillary passage time, without altering superior temporal arterial or venous diameters (Harris et al. 1995). These
results suggest enhanced perfusion of the
retina. In a subsequent comparison of timolol and betaxolol in normal tension
glaucoma, there was a dissociation between IOP alterations and blood flow
changes, with timolol (but not betaxolol)
significantly lowering IOP, while betaxolol (but not timolol) increasing end-diastolic velocities and reducing resistance indices in the average of four different
retrobulbar vessels (Harris et al. 1995).
These results suggest that betaxolol can
enhance ocular perfusion via a non-IOP
lowering mechanism.
Another pharmaceutical agent that
can enhance ocular perfusion is topical
dorzolamide. In 11 healthy subjects, dorzolamide reduced IOP decreased arteriovenous passage time, and increased
macular and superficial optic nerve head
capillary transit velocities, without altering the diameter or the superior temporal artery or vein. It did not alter flow
velocities or resistance indices in four
retrobulbar vessels on color Doppler imaging (Harris et al. 1996). Similar results
have been obtained in studies involving
subjects with normal tension glaucoma,
with a 24% decrease in retinal arteriovenous passage time after dorzolamide
treatment despite no significant alteration
in retrobulbar flow velocities (Harris et
al. 1996).
Another category of agents includes
calcium channel blockers. In one study,
there was a correlation between the mean
visual field defect and central retinal artery resistance index in 14 subjects with
normal tension glaucoma who were
treated with calcium channel blockers
(Pillanat et al. 1994, 1995). This study
also showed that those patients with the
greatest initial central retinal artery resistance index experienced the greatest improvement in mean visual field defect
(Pillanat et al. 1994, 1995). Another
study showed an improvement in mean
contrast sensitivity and alterations in
ophthalmic artery peak systolic and enddiastolic velocities in 16 of 21 normal tension glaucoma patients treated with oral
nifedipine (Harris et al. 1997). These
findings corroborate the idea that ocular
perfusion and ophthalmic visual function
are correlated.
Consequently, it is apparent that modu-
lation of ocular blood flow is possible
using currently available topical ophthalmics (glaucoma therapy) and systemic
agents (calcium channel antagonists).
These agents may improve visual function
as measured by contrast sensitivity and
visual field assessment. Further study of
these agents in nonexudative age-related
macular degeneration is warranted.
Conclusion
In summary, there appears to be a specific derangement in the choroidal circulation in patients with AMD. Specifically,
vascular defects have been identified in
both nonexudative and exudative AMD
patients using fluorescein angiographic
methods, laser Doppler flowmetry, indocyanine green angiography, and color
Doppler imaging. Although these studies
lend some support to the vascular pathogenesis of AMD, it is not possible to determine if the choroidal perfusion abnormalities play a causative role in nonexudative AMD, if they are simply an
association with another primary alteration, such as a primary RPE defect or a
genetic defect at the photoreceptor level,
or if they are more strongly associated
with one particular form of this heterogeneous disease. Specific issues to be addressed in future studies include correlation of perfusion defects with various
subtypes of nonexudative AMD, further
correlation of perfusion defects with severity of the disease process within these
subtypes, correlation of perfusion defects
with disease progression, and assessment
of currently-available vasoactive pharmaceutics in improving macular perfusion
and function in nonexudative AMD or to
slow disease progression in this disorder.
Acknowledgement
Supported, in part, by grants from Research to
Prevent Blindness, Inc.; and N.I.H. grant
EY10801. Dr. Ciulla is a recipient af a career
development award from Research to Prevent
Blindness, Inc., New York.
References
Allikmets R, Shroyer N, Singh N, Seddon J,
Lewis R, Bernstein P, Peiffer A, Zabriskie N,
Li Y, Hutchinson A, Dean M, Lupski J &
Leppert M (1997): Mutation of the Stargardt disease gene (ABCR) in age-related
macular degeneration. Science 277: 1805–
1807.
Amin R, Puklin J & Frank R (1994): Growth
factor localization in choroidal neovascular
membranes of age-related macular degeneration. Invest Ophthalmol Vis Sci 35:
3178–3188.
Boker T, Fang T & Steinmetz R (1993). Refractive error and choroidal perfusion characteristics in patients with choroidal neovascularization and age-related macular degeneration. Ger J Ophthalmol 2: 10–13.
Bone RA, Landrum JT, Hime GW, Cains A &
Zamor J (1993): Stereochemistry of the human macular carotenoids. Invest Ophthalmol Vis Sci 34: 2033–2040.
Bone RA, Landrum JT & Tarsis SL (1985):
Preliminary identification of the human
macular pigment. Vision Res 25: 1521–1525.
Bressler N, Bressler S & Fine S (1988): Agerelated
macular
degeneration.
Surv
Ophthalmol 32: 375–413.
Chen J, Fitzke F, Pauleikhoff D & Bird A
(1992): Functional loss in age-related
Bruch’s membrane change with choroidal
perfusion defect. Invest Ophthalmol Vis Sci
33: 334–340.
Chen J, Graham S, Nakayama M, Zhu R, Jin
K, Stetler R & Simon R (1997): Apoptosis
repressor genes Bcl-2 and Bcl-x-long are expressed in the rat brain following global
ischemia. J Cereb Blood Flow Metab 98: 2–
10.
Christen W, Glynn R, Manson J, Ajani U &
Buring J (1996): A prospective study of cigarette smoking and risk in age-related macular degeneration in men. JAMA 276: 1147–
1151.
Ciulla TA, Curran-Celentano J, Cooper DA,
Hammond BR Jr., Danis RP, Pratt LM,
Riccardi KA & Filloon TG (2000): Macular
pigment optical density in a Midwestern
sample. Ophthalmology (in press).
Ciulla TA, Danis RP & Harris A (1998): Agerelated macular degeneration: a review of experimental treatments. Surv Ophthalmol 43:
134–146.
Ciulla TA, Harris A, Chung HS, Danis RP,
Kagemann L, Martin BJ & Garzozi H
(2000): Choroidal perfusion is delayed in
non-exudative age-related macular degeneration. Ophthalmology (in press).
Ciulla TA, Harris A, Chung HS, Danis RP,
Kagemann L, McNulty L, Pratt LM & Martin BJ (1999): Color Doppler imaging discloses reduced ocular blood flow velocities
in nonexudative age-related macular degeneration. Am J Ophthalmol 128: 75–80.
Ciulla TA, Harris A & Danis RP (1998): Presumed macular choroidal watershed vascular filling, choroidal neovascularization, and
systemic vascular disease in patients with
age-related macular degeneration [letter;
comment]. Am J Ophthalmol 126: 153–155.
Cruickshanks K, Klein R & Klein B (1993):
Sunlight and age-related macular degeneration. The Beaver Dam Eye Study. Arch
Ophthalmol 111: 514–518.
113
De La Paz M, Guy V, Abou-Donia S, Heinis
R, Bracken B, Vance J, Gilbert J, Gass J,
Haines J & Pericak-Vance M (1999): Analysis of the Stargardt disease gene (ABCR) in
age-related macular degeneration. Ophthalmology 106: 1531–1536.
De La Paz MA, Pericak-Vance MA, Lennon F,
Haines JL & Seddon JM (1997): Exclusion
of TIMP3 as a candidate locus in age-related
macular degeneration. Invest Ophthalmol
Vis Sci 38: 1060–1065.
Del Priore L, Hornbeck R, Kaplan H, Jores Z,
Valentino T, Mosinger-Ogilvie J & Swinn M
(1995): Debridement of the pig retinal pigment epithelium in vivo. Arch Ophthalmol
113: 939–944.
Del Priore L, Kaplan H, Hornbeck R, Jones
Z & Swinn M (1996): Retinal pigment epithelial debridement as a model for the
pathogenesis and treatment of macular degeneration. Am J Ophthalmol 122: 629–643.
Eagle RJ (1984): Mechanisms of maculopathy.
Ophthalmology 91: 613–625.
Faris R, Apte S, Olsen B, Iwata K & Milam A
(1997): Tissue inhibitor of metalloproteinases-3 is a component of Bruch’s membrane
of the eye. Am J Pathol 150: 323–328.
Freund K, Yannuzzi L & Sorenson J (1993):
Age-related macular degeneration and choroidal neovascularization. Am J Ophthalmol
115: 786–791.
Friedman E (1997): A hemodynamic model of
the pathogenesis of age-related macular degeneration. Am J Ophthalmol 124: 677–682.
Friedman E, Ivry M, Ebert E, Glynn R, Gragoudas ES & Seddon J (1989): Increased
scleral rigidity and age-related macular degeneration. Surv Ophthalmol 96: 104–108.
Friedman E, Krupsky S, Lane A, Oak S,
Friedman E, Egan K & Gragoudas E
(1995): Ocular blood flow velocity in age-related macular degeneration. Ophthalmology
102: 640–646.
Gillardon F, Lenz C, Waschke K, Krajewski S,
Reed J, Zimmerman M & Kuschinsky W
(1996): Altered expression of Bcl-2, Bcl-x,
Bax, and c-Fos colonizes with DNA fragmentation and ischemic cell damage following middle cerebral artery occlusion in rats.
Brain Res Mol Brain Res 40: 254–260.
Green W, McDonnell P & Yeo J (1985): Pathologic features of senile macular degeneration. Ophthalmology 92: 615–627.
Grunwald J, Hariprasad S, DuPont J, Maguire
M, Fine S, Brucker A, Maguire A & Ho A
(1998): Foveolar choroidal blood flow in
age-related macular degeneration. Invest
Ophthalmol Vis Sci 1998: 385–390.
Hammond BR Jr., Curran-Celentano J, Judd
S, Fuld K, Krinsky NI, Wooten BR &
Snodderly DM (1996): Sex differences in
macular pigment optical density: relation to
plasma carotenoid concentrations and dietary patterns. Vision Res 36: 2001–2012.
Hammond BR Jr., Fuld K & Snodderly DM
(1996): Iris color and macular pigment optical density. Exp Eye Res 62: 293–297.
Hammond BR Jr., Wooten BR & Snodderly
114
DM (1996): Cigarette smoking and retinal
carotenoids: implications for age-related
macular degeneration. Vision Res 36: 3003–
3009.
Harris A, Arend O, Arend S & Martin B
(1996): Effects of topical dorzolamide on
retinal and retrobulbar hemodynamics. Acta
Ophthalmol Scand 74: 569–572.
Harris A, Chung HS, Ciulla TA & Kagemann
L (1999): Progress in measurement of ocular
blood flow and relevance to our understanding of glaucoma and age-related macular degeneration. Prog Retin Eye Res 18: 669–
687.
Harris A, Ciulla TA, Kagemann L, Zarfait
D & Martin B (2000): Vasoprotection as neuroprotection for the optic nerve. Eye 14:
473–475.
Harris A, Evans D, Cantor L & Martin B
(1997): Hemodynamic and visual function
effects of oral nifedipine in normal tension
glaucoma. Am J Ophthalmol 124: 296–302.
Harris A, Kagemann L, Chung H & Ciulla T
(1998): Ophthalmic Imaging and Diagnostics: The use of dye dilution curve analysis in
the quantification of indocyanine green angiograms of the human choroid. Ophthalmology Clinics of North America, St Louis,
W. B. Saunders.
Harris A, Shoemaker J, Burgoyne J, Weinland
M, Sponsel W & Cantor L (1995): The acute
effect of topical beta-adrenergic antagonists
on normal perimacular hemodynamics. J
Glaucoma 4: 36–40.
Harris A, Spaeth G, Sergott R, Katz L, Cantor
L & Martin B (1995): Retrobulbar arterial
hemodynamic effects of betaxolol and timolol in normal-tension glaucoma. Am J
Ophthalmol 120: 168–175.
Henkind P & Gartner S (1983): The relationship between retinal pigment epithelium and
the choriocapillaris. Trans Ophthalmol Soc
UK 103: 444–447.
Hyman L, He O, Grimson R, Oden N, Schachat A, Leske M & a T A-r MPS Group
(1992): Risk factors for age-related maculopathy. Invest Ophthalmol Vis Sci (Suppl) 33:
548.
Ishibashi T, Inomata H, Sakamoto T & Ryan
S (1995): Pericytes of newly formed vessels
in experimental subretinal neovascularization. Arch Ophthalmol 113: 227–231.
Ishibashi T, Miki K, Sorgente N, Patterson
R & Ryan S (1985): Effects of intravitreal
administration of steroids on experimental
subretinal neovascularizalion in the subhuman primate. Arch Ophthalmol 103: 708–
711.
Ishibashi T, Miller H, Orr G, Sorgente N &
Ryan S (1987): Morphologic observations
on experimental subretinal neovascularization in the monkey. Invest Ophthalmol Vis
Sci 28: 1116–1130.
Kamei M & Holleyfield J (1999): TIMP-3 in
Bruch’s membrane: change during aging and
in age-related macular degeneration. Invest
Ophthalmol Vis Sci 40: 2367–2375.
Katz M, Parker K, Handelman G, Bramel T &
Ea D (1982): Effects of antioxidant nutrient
deficiency on the retina and retinal pigment
epithelium of albino rats: A light and electron microscopic study. Exp Eye Res 34:
339–369.
Klaver C, Kliffen M, van Duijn C, Hofman A,
Cruts M, Grobbee D, van Broeckhoven C &
de Jong P (1998): Genetic association of
apoliprotein E with age-related macular degeneration. Am J Hum Genet 63: 200–206.
Klein R, Klein B & Linton K (1992): Prevalence of age-related maculopathy. The Beaver Dam Eye Study. Ophthalmology 99:
933–943.
Korte G, Reppucci V & Henkind P (1984):
RPE destruction cause choriocapillary atrophy. Invest Ophthalmol Vis Sci 25: 1135–
1145.
Kvanta A (1995): Expression and regulation of
vascular endothelial growth factor in choroidal fibroblasts. Curr Eye Res 14: 1015–
1020.
Leib S, Kim Y, Chow L, Sheldon T & Tauber
M (1996): Reactive oxygen intermediates
contribute to necrotic and apoptotic neuronal injury in an infant rat model of bacterial meningitis due to group B streptococci. J Clin Invest 98: 2632–2639.
Leibowitz H, Krueger D, Maunder L, Milton
R, Kini M, Kahn H, Nickerson R, Pool J,
Colton T, Ganley J, Loewenstein J & Dawber T (1980): The Framingham Eye Study
monograph: An ophthalmological and epidemiological study of cataract, glaucoma,
diabetic retinopathy, macular degeneration,
and visual acuity in a general population of
2631 adults, 1973–1975. Surv Ophthalmol
24: 335–610.
Lopez PF, Sippy BD, Lambert HM, Thach
AB & Hinton DR (1996): Transdifferentiated retinal pigment epithelial cells are immunoreactive for vascular endothelial
growth factor in surgically excised age-related macular degeneration-related choroidal neovascular membranes. Invest
Ophthalmol Vis Sci 37: 855–868.
Macaya A (1996): Apoptosis in the nervous
system. Rev Neurol 24: 1356–1360.
Malinow MR, Feeney-Burns L, Peterson LH,
Klein ML & Neuringer M (1980): Diet-related macular anomalies in monkeys. Invest
Ophthalmol Vis Sci 19: 857–863.
Mares-Perlman J, Brady W, Klein R, Klein B,
Bowen P, Stacewicz-Sapuntzakis M & Palta
M (1995): Serum antioxidants and age-related macular degeneration in a populationbased case-control study. Arch Ophthalmol
113: 1518–1523.
Miller H, Miller B, Ishibashi T & Ryan S
(1990): Pathogenesis of laser-induced choroidal subretinal neovascularization. Invest
Ophthalmol Vis Sci 31: 899–908.
Moisseiev J, Alhalel A, Masuri R & Treister G
(1995): The impact of the macular photocoagulation study results on the treatment of
exudative age-related macular degeneration.
Arch Ophthalmol 113: 185–189.
MPS (1982): Argon laser photocoagulation for
senile macular degeneration. Results of a
randomized clinical trial. Arch Ophthalmol
100: 912–918.
MPS (1986): Argon laser photocoagulation for
neovascular maculopathy. Three-year results
from randomized clinical trials. Arch
Ophthalmol 104: 694–701.
MPS (1991): Argon laser photocoagulation for
neovascular maculopathy. Five-year results
from randomized clinical trials. Arch
Ophthalmol 109: 1109–1114.
MPS (1993): Five-year follow-up of fellow eyes
of patients with age-related macular degeneration and unilateral extrafoveal choroidal neovascularization. Arch Ophthalmol
111: 1189–1199.
Nasir M, Sugino I & Zarbin M (1997): Decreased choriocapillaris perfusion following
surgical excision of choroidal neovascular
membranes in age-related macular degeneration. Br J Ophthalmol 81: 481–489.
Neuringer M, Klein M, Snodderly M, Hohnson E, Feeney-Burns L & Kittleson C
(1999): Effects of carotenoid depletion and
N-3 fatty acid status on macular defects in
Rhesus monkeys (ARVO abstract). Invest
Ophthalmol Vis Sci (suppl) 40: 164.
Nickells R (1996): Retinal ganglion cell death
in glaucoma: the how, the why, and the
maybe. J Glaucoma 5: 345–356.
Nishimura T, Goodnight R, Prendergast R &
Ryan S (1990): Activated macrophages in
experimental subretinal neovascularization.
Ophthalmologica 200: 39–44.
Ohkuma H & Ryan S (1982): Vascular casts of
experimental subretinal neovascularization
in monkeys: a preliminary report. Jpn J
Ophthalmol 26: 150–158.
Ohkuma H & Ryan S (1983): Experimental subretinal neovascularization in the monkey.
Permeability of new vessel. Arch Ophthalmol 101: 1102–1110.
Ohkuma H & Ryan S (1983): Vascular casts of
experimental subretinal neovascularization
in monkeys. Invest Ophthalmol Vis Sci 24:
481–490.
Pauleikhoff D, Chen J, Chisholm I & Bird A
(1990): Choroidal perfusion abnormality
with age-related Bruch’s membrane change.
Am J Ophthalmol 109: 211–217.
Piguet B, Palmvang I, Chisholm I, Minassian
D & Bird A (1992): Evolution of age-related
macular degeneration with choroidal perfusion abnormality. Am J Ophthalmol 113:
657–663.
Pillunat L, Lang G & Harris A (1994): The
visual response to increased ocular blood
flow in normal pressure glaucoma. Surv
Ophthalmol 38: S139-S145.
Pillunat L, Lang G & Harris A (1995): Ocular
carbon dioxide reactivity and calcium channel blockers in normal tension glaucoma.
Glaucoma, Ocular Blood Flow and Drug
Treatment. S. M. Drance. Amsterdam, Kugler: 67–71.
Pollack J, Del Priore L, Smith M, Feiner M &
Kaplan H (1996): Postoperative abnormalities of the choriocapillaris in exudative age-
related macular degeneration. Br J Ophthalmol 80: 314–318.
Quigley H, Nickells R, Kerrigan L, Pease M,
Thibault D & Zack D (1995): Retinal ganglion cell death in experimental glaucoma
and after axotomy occurs by apoptosis. Invest Ophthalmol Vis Sci 36: 774–786.
Reddy VM, Zamora RL & Kaplan HJ (1995):
Distribution of growth factors in subfoveal
neovascular membranes in age-related
macular degeneration and presumed ocular
histoplasmosis syndrome. Am J Ophthalmol
120: 291–301.
Remulla J, Gaudio A, Miller S & Sandberg M
(1995): Foveal electroretinograms and choroidal perfusion characteristics in fellow eyes
of patients with unilateral neovascular agerelated macular degeneration. Br J Ophthalmol 79: 558–561.
Ross R, Barofsky J, Cohen G, Baber W, Palao
S & Gitter K (1998): Presumed macular choroidal watershed vascular filling, choroidal
neovascularization, and systemic vascular
disease in patients with age-related macular
degeneration. Am J Ophthalmol 125: 71–80.
Ryan S (1979): The development of an experimental model of subretinal neovascularization in disciform macular degeneration.
Trans Am Ophthalmol Soc 77: 707–745.
Ryan S (1980): Subretinal neovascularization.
Natural history of an experimental model.
Arch Ophthalmol 100: 1804–1809.
Sarks SH (1976): Ageing and degeneration in
the macular region: a clinico-pathological
study. Br J Ophthalmol 60: 324–341.
Schalch W (1992): Carotenoids in the retina –
a review of their possible role to prevent or
limit damage caused by light and oxygen.
Free Radicals and Aging 62: 280–298.
Seddon J, Ajani U, Sperduto R, Hiller R, Blair
N, Burton T, Farber M, Gragoudas E, Haller J, Miller D et al. (1994): Dietary carotenoids, vitamins A, C, and E, and advanced
age-related macular degeneration. Eye Disease Case-Control Study Group [see comments] [published erratum appears in JAMA
1995 Feb 22;273(8):622]. JAMA 272: 1413–
1420.
Seddon J & Hennekens C (1994): Vitamins,
minerals, and macular degeneration: Promising but unproven hypotheses. Arch
Ophthalmol 112: 176–178.
Seddon J, Willet W, Speizer F & Hankinson S
(1996): A prospective study of cigarette
smoking and age-related macular degeneration in women. JAMA 276: 1141–1146.
Snodderly DM, Auran JD & Delori FC (1984):
The macular pigment. II. Spatial distribution in primate retinas. Invest Ophthalmol Vis Sci 25: 674–685.
Souied E, Benlian P, Amouyel P, Feingold J,
Lagarde J, Munnich A, Kaplan J, Coscas
G & Soubrane G (1998): The epsilon4 allele
of the apolipoprotein E gene as a potential
protective factor for exudative age-related
macular degeneration. Am J Ophthalmol
125: 353–359.
Spraul C & Grossniklaus H (1997): Character-
istics of drusen and Bruch’s membrane in
postmortem eyes with age-related macular
degeneration. Arch Ophthalmol 115: 267–
273.
Steen B, Sejersen S, Berglin L, Seregard S &
Kvanta A (1998): Matrix metalloproteinases
and metalloproteinase inhibitors in choroidal neovascular membranes. Invest
Ophthalmol Vis Sci 39: 2194–2200.
Stone E, Lotery A, Munier F, Heon E, Piguet
B, Guymer R, Vandenburgh K, Cousin P,
Nishimura D, Swiderski R, Silvestri G,
Mackey D, Hageman G, Bird A, Sheffield
V & Schorderet D (1999): A single EFEMP1
mutation associated with both Malattia Leventinese and Doyne honeycomb retinal
dystrophy. Nat Genet 22: 199–202.
Stone E, Webster A, Vandenburgh K, Streb L,
Hockey R, Lotery A & Sheffield V (1998):
Allelic variation in ABCR associated with
Stargardt disease but not age-related macular degeneration (letter). Nat Genet 20: 328–
329.
Takeuchi M, Itagaki T, Takahashi K, Ohkuma
H & Uyama M (1993): Changes in the intermediate stage of retinal degeneration after
intravitreal injection of ornithine. Nippon
Ganka Gakkai Zasshi 1: 17–28.
Tielsch J, Javitt J, Coleman A, Katz J &
Sommer A (1995): The prevalence of blindness and visual impairment among nursing
home residents in Baltimore [see comments].
N Engl J Med 332: 1205–1209.
Weber B, Vogt G, Pruett R, Stohr H & Felbor
U (1994): Mutations in the tissue inhibitor
of metalloproteinases-3 (TIMP3) in patients
with Sorsby’s fundus dystrophy. Nat Genet
4: 352–356.
Weber B, Vogt G, Wolz W, Ives E & Ewing C
(1994): Sorsby’s fundus dystrophy is genetically linked to chromosome 22q13-qter. Nat
Genet 7: 158–161.
Weiter JJ, Delori F & Dorey CK (1988): Central sparing in annular macular degeneration. Am J Ophthalmol 106: 286–292.
West S, Vitale S, Hallfrisch J (all names, please)
(1994): Antioxidants and supplements: Protective for age-related macular degeneration?
Arch Ophthalmol 112: 222–227.
Young R (1987): Pathophysiology of age-related macular degeneration. Surv Ophthalmol 31: 291–306.
Zhao J, Frambach D, Lee P, Lee M & Lopez
P (1995): Delayed macular choriocapillary
circulation in age-related macular degeneration. Int Ophthalmol 19: 1–12.
Received on August 18th, 2000.
Accepted on August 25th, 2000.
Corresponding author:
Alon Harris, Ph.D.
702 Rotary Circle, IUMC
Indianapolis, IN, 46260
USA
Phone: 317 278 0134
115
Download