ArticlePDF Available

Decreased thickness and integrity of the macular elastic layer of Bruch’s membrane correspond to the distribution of lesions associated with age-related macular degeneration

Authors:

Abstract and Figures

Age-related macular degeneration (AMD) is a leading cause of blindness in the elderly. In its severest form, choroidal neovessels breach the macular Bruch's membrane, an extracellular matrix compartment comprised of elastin and collagen laminae, and grow into the retina. We sought to determine whether structural properties of the elastic lamina (EL) correspond to the region of the macula that is predilected toward degeneration in AMD. Morphometric assessment of the macular and extramacular regions of 121 human donor eyes, with and without AMD, revealed a statistically significant difference in both the integrity (P < 0.0001) and thickness (P < 0.0001) of the EL between the macular and extramacular regions in donors of all ages. The EL was three to six times thinner and two to five times less abundant in the macula than in the periphery. The integrity of the macular EL was significantly lower in donors with early-stage AMD (P = 0.028), active choroidal neovascularization (P = 0.020), and disciform scars (P = 0.003), as compared to unaffected, age-matched controls. EL thickness was significantly lower only in individuals with disciform scars (P = 0.008). The largest gaps in macular EL integrity were significantly larger in all categories of AMD (each P < 0.0001), as compared to controls. EL integrity, thickness, and gap length in donors with geographic atrophy did not differ from those of controls. These structural properties of the macular EL correspond spatially to the distribution of macular lesions associated with AMD and may help to explain why the macula is more susceptible to degenerative events that occur in this disease.
Content may be subject to copyright.
Molecular Pathogenesis of Genetic and Inherited Diseases
Decreased Thickness and Integrity of the Macular
Elastic Layer of Bruch’s Membrane Correspond to
the Distribution of Lesions Associated with Age-
Related Macular Degeneration
N.H. Victor Chong,*
Jason Keonin,*
Phil J. Luthert,
Christina I. Frennesson,
§
David M. Weingeist,* Rachel L. Wolf,*
Robert F. Mullins,* and Gregory S. Hageman*
From the Department of Ophthalmology and Visual Sciences,*
The University of Iowa Center for Macular Degeneration, The
University of Iowa, Iowa City, Iowa; the Department of
Ophthalmology,
King’s College, London, United Kingdom; The
Institute of Ophthalmology,
University College London and
Moorfields Eye Hospital, London, United Kingdom; and the
Department of Neuroscience and Locomotion,
§
Division of
Ophthalmology, Linkoping University, Linkoping, Sweden
Age-related macular degeneration (AMD) is a leading
cause of blindness in the elderly. In its severest form,
choroidal neovessels breach the macular Bruch’s
membrane, an extracellular matrix compartment
comprised of elastin and collagen laminae, and grow
into the retina. We sought to determine whether
structural properties of the elastic lamina (EL) corre-
spond to the region of the macula that is predilected
toward degeneration in AMD. Morphometric assess-
ment of the macular and extramacular regions of 121
human donor eyes, with and without AMD, revealed a
statistically significant difference in both the integrity
(P<0.0001) and thickness (P<0.0001) of the EL
between the macular and extramacular regions in
donors of all ages. The EL was three to six times
thinner and two to five times less abundant in the
macula than in the periphery. The integrity of the
macular EL was significantly lower in donors with
early-stage AMD (P0.028), active choroidal neovas-
cularization (P0.020), and disciform scars (P
0.003), as compared to unaffected, age-matched con-
trols. EL thickness was significantly lower only in
individuals with disciform scars (P0.008). The larg-
est gaps in macular EL integrity were significantly
larger in all categories of AMD (each P<0.0001), as
compared to controls. EL integrity, thickness, and
gap length in donors with geographic atrophy did not
differ from those of controls. These structural prop-
erties of the macular EL correspond spatially to the
distribution of macular lesions associated with AMD
and may help to explain why the macula is more
susceptible to degenerative events that occur in this
disease. (Am J Pathol 2005, 166:241–251)
Bruch’s membrane is a stratified extracellular matrix com-
plex that lies between the retinal pigment epithelium
(RPE) and the choroidal capillary bed, or choriocapillaris.
It is comprised of two collagen-rich layers, referred to as
the inner and outer collagenous layers, that flank a cen-
tral domain of elastin and elastin-associated proteins.
1,2
A number of age-related changes have been described
in Bruch’s membrane,
3–23
the most prominent of which
are drusen and basal laminar deposits.
24–30
In addition,
increases in thickness, enhanced basophilia and Su-
danophilia, accumulation of membranous debris, de-
creases in hydraulic conductivity, and fragmentation and
calcification of Bruch’s membrane, have been described.
These age-related alterations in Bruch’s membrane
could lead to a loss of the normal function of Bruch’s
membrane and promote degenerative changes in the
aging eye. Various lines of evidence suggest that Bruch’s
membrane functions as a physical barrier to the egress of
cells and vessels from the choroid into the sub-RPE and
subretinal spaces. Disruption of, or damage to, this bar-
rier is associated with loss of vision in a variety of ocular
Supported in part by the National Institutes of Health (grants EY11515 to
G.S.H. and EY014563 to R.F.M.); The Ruth and Milton Steinbach Fund,
Inc. (to G.S.H.); a Research to Prevent Blindness Lew R. Wasserman Merit
Award (to G.S.H.); and The University of Iowa Center on Aging (grant
AG00214 to R.F.M.).
Accepted for publication September 16, 2004.
Address reprint requests to Gregory S. Hageman, Ph.D., Department of
Ophthalmology and Visual Sciences, Center for Macular Degeneration,
The University of Iowa, 11190E PFP, 200 Hawkins Dr., Iowa City, IA 52240.
E-mail: gregory-hageman@uiowa.edu.
American Journal of Pathology, Vol. 166, No. 1, January 2005
Copyright © American Society for Investigative Pathology
241
diseases, often resulting from the growth of new blood
vessels—termed choroidal neovascular membranes
(CNVM)—from the choroid into the sub-RPE and/or sub-
retinal spaces. CNVM formation occurs in a number of
macular degenerative diseases, including age-related
macular degeneration (AMD), the leading cause of irre-
versible blindness in the developed world.
31
Moreover,
CNVM formation can be induced experimentally by ther-
mal damage to Bruch’s membrane in normal monkey
eyes after laser photocoagulation
32
and potentially, after
laser treatment of human CNVM.
33
Interestingly, extra-
macular treatment is relatively ineffective, suggesting that
the macula is inherently susceptible to choroidal neovas-
cularization (CNV).
34
AMD-associated CNVM formation occurs in 4% of
the population older than the age of 75
35
and is primarily
restricted to the macula. It accounts for 90% of severe
visual loss associated with AMD, even though only 10
to 15% of individuals with AMD develop CNV. The macula
is a unique 6-mm-diameter region of the posterior pole
that lies directly in the visual axis of human and nonhu-
man primates (Figure 1A). This region of the retina de-
velops postnatally and subserves fine acuity vision. The
reason for the increased susceptibility of the macula to
degeneration and CNVM formation in primates has not
been elucidated.
We have speculated that one possible explanation for
the tendency of new vessels to breach the macular
Bruch’s membrane, as opposed to the extramacular re-
gions, might be that there are regional differences in the
structure and/or composition of Bruch’s membrane. It is
likely that the integrity and nature of its collagen and
elastin components primarily dictate the basic structural
and functional properties of Bruch’s membrane. Hence,
one might predict that the disruption of one or more
layers of Bruch’s membrane might precede CNV in AMD.
Accordingly, we recently observed what we interpreted
to be robust differences in the integrity and thickness of
the elastic lamina (EL) of Bruch’s membrane between the
macular and extramacular regions in a series of human
eyes. To evaluate further the topographical variation in
the thickness and continuity of the EL, we assessed these
parameters in a larger series of normal, unaffected donor
eyes and compared them to those obtained from donors
with early and late stages of AMD.
The data collected herein show that the macular EL is
substantially thinner and more porous than that in the
extramacular region. This information has lead to the
development of a working hypothesis that the structural
attributes of the macular EL of Bruch’s membrane may be
related to, at least in part, the predilection of this region to
degeneration in AMD, other macular dystrophies, and
other conditions characterized by CNVM formation.
Materials and Methods
Human Donor Eyes and Morphological
Procedures
The 121 human eyes used in this study were obtained
from MidAmerica Transplant Services (St. Louis, MO), the
Iowa Lions Eye Bank (Iowa City, IA), the Heartland Eye
Bank (Columbia, MO), the Central Florida Lions Eye and
Tissue Bank (Tampa, FL), and the Virginia Eye Bank
(Norfolk, VA) after informed consent. Institutional Review
Board committee approval for the use of human donor
tissues was obtained from Human Subjects Committees
at St. Louis University and the University of Iowa. All eyes
were processed within 4 hours of death. The gross patho-
logical features, as well as the corresponding fundus
photographs and angiograms when available, of all eyes
Figure 1. Ocular regions used in this study. A: Color funduscopic photograph of the left eye of a middle-aged individual without any clinical signs of AMD. The
macula (dashed circle) is centered on the fovea (arrow) and lies within the major retinal vessels (arrowheads) that emanate from the optic nerve head (ONH).
The superior (S), inferior (I), temporal (T), and nasal (N) axes are marked for orientation. B: Diagrammatic representation of the distribution of the 2-mm-diameter
retinal-choroidal-scleral punches collected from a 14-year-old donor. The solid black circle depicts the location of the optic nerve head, the curved black lines
emanating from the optic disk represent the major retinal vessels, and the blue circle denotes the punch that was centered on the fovea. The remaining circles
represent serial 2-mm-diameter punches taken along the superior, inferior, temporal, and nasal axes. C: Illustration showing the macular and extramacular
(mid-peripheral) sites (circles) from which 4-mm-diameter punches were collected for EL thickness and integrity measurements in the large set of AMD and
unaffected donors. Landmarks are the same as those described in B.
242 Chong et al
AJP January 2005, Vol. 166, No. 1
in this repository were read and classified by retinal spe-
cialists. Fundi were classified according to a modified
version of the International AMD grading system.
36
For
this investigation, donors were placed into one of four
categories: young unaffected (62 years), age-matched
unaffected (62 years), early-stage AMD (the youngest
was 62 years old), and late-stage AMD. Late-stage AMD
donors were subdivided into those with 1) geographic
atrophy (GA), 2) active CNVMs, and 3) disciform scars
preceded clinically by CNVM (CNV/DS). Note that some
individuals refer to early-stage AMD as age-related
maculopathy, or ARM; this term will not be used herein.
Donors were classified as unaffected if they had no mac-
roscopic or funduscopic signs of macular pathology or
any ophthalmic history of AMD. Early AMD donors were
defined based on an ophthalmic history of AMD and the
presence of significant numbers of macular drusen, pig-
ment disruption, and/or other clinical signs of early AMD.
Polyclonal antisera directed against tropoelastin
(PCAB 94, gift from Dr. Robert Mecham, Washington
University, St. Louis, MO; PR398, Elastin Products,
Owensville, MO), as well as monoclonal antibodies di-
rected against bovine elastin (clone BA-4, Sigma, St.
Louis, MO; MM436, Elastin Products) and human aortic
-elastin (PR533, Elastin Products) were used to examine
the EL of Bruch’s membrane. For experiments using Elas-
tin Products’ antibodies, antigens were retrieved accord-
ing to the manufacturer’s instructions. Reactivity of anti-
bodies with the EL of Bruch’s membrane was analyzed in
a series of young (10 years), middle-aged (20 to 60
years), and aged (60 years) donors. Posterior poles, or
wedges of posterior poles spanning between the ora
serrata and the macula, were fixed in 4% (para)formal-
dehyde in 100 mmol/L sodium cacodylate, pH 7.4, as
described previously.
29
After 2 to 4 hours of fixation, eyes
were transferred to 100 mmol/L sodium cacodylate and
rinsed (3 10 minutes), infiltrated, and embedded in
acrylamide. These tissues were subsequently embedded
in OCT, snap-frozen in liquid nitrogen, and stored at
80°C. In addition, unfixed posterior poles, or wedges
thereof, were embedded directly in OCT, without acryl-
amide infiltration or embedment. Both fixed and unfixed
tissues were sectioned to a thickness of 6 to 8
mona
cryostat. Immunolabeling was performed as described
previously,
29,30
using Alexa 488-conjugated secondary
antibodies (Molecular Probes, Eugene, OR). Adjacent
sections were incubated with secondary antibody alone,
to serve as negative controls. Some immunolabeled
specimens were viewed by confocal laser-scanning mi-
croscopy.
37
These cellular nuclei in these sections were
counterstained with TO-PRO-3 (Molecular Probes).
Ocular tissues used for transmission electron micro-
scopical studies were fixed by immersion fixation in one-
half strength Karnovsky’s fixative, within 4 hours of death,
for a minimum of 24 hours. Trephine-punched specimens
(see below) were fixed, transferred to 100 mmol/L sodium
cacodylate buffer, pH 7.4, and subsequently dehydrated,
embedded in epoxy resin, sectioned, and photographed,
as described previously.
38,39
Morphometric Analyses
To establish the baseline structural and topographical
characteristics of the EL, irrespective of aging or AMD,
three separate analyses were performed. In the first anal-
ysis, baseline topographic data were collected from a
series of oriented, 2-mm-diameter, full-thickness punches
of RPE-choroid-sclera that were collected using a tre-
phine punch. These punches were taken at 2-mm inter-
vals from the fovea to the ora serrata in the temporal,
nasal, inferior, and superior quadrants in an eye from a
14-year-old donor and subsequently prepared for elec-
tron microscopy (Figure 1B). The average thickness and
integrity of the EL in each of these punches were mea-
sured. A second series of 2-mm-diameter punches de-
rived from two additional donors aged 7 and 25 were
measured (these data were similar to those of the 14-
year-old and are not depicted in this article).
In the second analysis, measurements of EL thickness
and integrity were made at two defined locations, 1 to 2
mm and 12 to 13 mm, from the foveal center, in the
infero-temporal quadrant, in a series of donors (Figure
1C). Fifty-six eyes from 56 unaffected (non-AMD) donors,
ranging in age from 6 hours after birth to 96 years of age
(mean age, 51.4 years), were used in this analysis. At
least five donor eyes from each decade of life were
included in this analysis. Oriented, 4-mm-diameter, full-
thickness punches of RPE-choroid-sclera were taken us-
ing a trephine punch and prepared for electron micros-
copy, as described above.
In the third analysis, similar measurements of macular
EL thickness and integrity were made from punches
taken from 64 eyes derived from 64 human AMD donors,
ranging in age from 62 to 99 years of age (Figure 1C).
These included 24 donors with macular drusen, pigment
disruption, and other signs of early-stage AMD; 15
donors with GA; 9 donors with active, patent CNVMs;
and 16 donors with disciform scars (DS) that were
preceded by documented CNVMs or in which a CNVM
was present in the second eye. The mean ages of
donors in these four categories were 79.0, 82.8, 83.7,
and 88.2 years, respectively.
Outcome Measures
Four random photographic images were taken from each
punched specimen using a JEOL JEM 1220 microscope
(JEOL USA Inc., Peabody, MA), as described previous-
ly.
38
Images were collected at 5000 actual magnifica-
tion for the first analysis (see above) and 2500 actual
magnification for the second and third analyses (see
above). The thickness of the EL of Bruch’s membrane
was measured at 20 points (five equal-distance points
from each of the four images per specimen) using a
micrometer. The average value of the 20 points was used
in the statistical analyses. The integrity of the EL was
defined as the total length of visually detectable elastin
divided by the overall length of visible Bruch’s membrane
within each of the four images. Thus, 100% integrity
represented a fully intact (nonporous) EL and 0% integrity
represented complete absence of elastin. The average
Topographical Variation in Bruch’s Membrane 243
AJP January 2005, Vol. 166, No. 1
integrity value derived from the four images of each
punch was used for statistical analyses. The largest, or
maximum, gap length (ie, discontinuity in the EL) present
within the elastin lamina from the four micrographs used
for each donor was identified and measured. The largest
gap length from all donors in any given category was
averaged (Figure 2F).
Statistical analyses were performed using Microsoft
Excel (Microsoft Inc., Redmond, WA) and S-Plus Statisti-
cal Packages (Insightful Corp., Seattle, WA). The Mann-
Whitney U-test (Wilcoxon rank-sum test) was used to
compare data series and linear regression analysis was
used to explore age-related changes. To compare nor-
mal, unaffected donors to donors with AMD, statistical
analyses were performed using only the normal, unaf-
fected donors 61 years of age (23 unaffected, 23 early-
stage AMD, and 40 late-stage AMD total). The mean age
of normal donors 61 was 79.8 years; this mean age was
not significantly different from that of each of the three
groups of donors with AMD. Some data are represented
graphically in the form of box and whiskers plots. These
graphs show values for the median (bolded line within
box), the first and third quartiles as a solid box, and the
minimum and maximum observation range as whiskers.
Any extreme values, defined as those further than 1.5
the interquartile range from the median, are represented
as solitary, horizontal lines.
Results
Immunohistochemistry
The reactivity of anti-elastin antibodies with the macular
EL of Bruch’s membrane differed markedly from that of
extramacular regions in all donor eyes examined (Figure
2, A and B). Immunoreactive elastin was attenuated and
highly discontinuous in the maculas of most donors, as
compared to more peripheral regions where it was con-
tinuous and thick. This same pattern was observed with
all elastin antibodies tested. The differences between
these same structural attributes in the macular and ex-
tramacular regions are even more striking when viewed
Figure 2. Confocal (A,B) and transmission electron microscopical (C–F) images of the elastic layer of Bruch’s membrane. Images were collected from macular
(A,C,E) and extramacular (B,D,F) regions of Bruch’s membrane, as indicated in Figure 1. Aand B: Labeling with an antibody directed against tropoelastin in
a 78-year-old donor without macular disease. Tissues prepared for confocal microscopy were counterstained with TO-PRO-3 to visualize cellular nuclei (blue);
RPE-associated lipofuscin is autofluorescent (red). Note that the elastin immunoreactivity of the elastic layer (green) is far more robust in the extramacular region
(B) that it is in the macular region (A). The EL (arrows) in the macular region of an 82-year-old donor, depicted in Cand D, is thinner and contains more
numerous discontinuities than in the extramacular region. E: A higher magnification image of Bruch’s membrane directly adjacent to the foveal pit; the elastin
fibers in this region are very sparse and thin (arrow). When the extramacular EL is viewed en face (F), its porosity (asterisk) is evident. These discontinuities
contribute to the integrity and gap length values measured in this study. Original magnifications, 400 (A,B). Scale bar, 2
m(C,D).
244 Chong et al
AJP January 2005, Vol. 166, No. 1
by transmission electron microscopy (Figure 2, C and D).
In the center of the fovea, elastin was particularly sparse
and thin (Figure 2E). These data provided the impetus for
a more robust evaluation of these structural parameters in
a larger set of samples derived from donors with and
without a history of AMD.
EL Integrity and Thickness Maps Derived from a
Young Donor
The mean integrity of the EL measured in the series of
punches derived from a 14-year-old donor (Figure 1B) is
depicted in Figure 3, A and B. The mean integrity was
lowest (40%) in the central macula (fovea) and in-
creased rapidly, approaching 80% at the main retinal
vascular arcades (8mmand8 mm) in the temporal,
superior, and inferior regions. In the nasal quadrant, the
integrity was lowest around the optic disk, especially on
the nasal aspect, and rapidly increased in regions ap-
proaching 8 to 10 mm from the fovea, just outside the
major vascular arcades. The mean thickness values ex-
hibited a similar distribution, except there was not a sig-
nificant fovea-associated dip (Figure 3, C and D). A cal-
culated color-coded schematic map of the mean EL
integrity of this eye is depicted in Figure 4A. In general,
the area of the fundus with the thinnest and most porous
EL corresponds spatially to the same region where the
majority of lesions associated with AMD are manifest
(Figure 4, B and C).
EL Integrity and Thickness in the Macular and
Extramacular Regions of Donors without AMD
Scatter plots for the mean integrity and thickness of the
EL of Bruch’s membrane in a series of normal, unaffected
human donors are depicted in Figure 5. The mean integ-
rity value of the EL in this series of normal human donors
was 37.2% (SD 7.9%) in the macula and 92.3% (SD
4.7%) in the extramacula (Figure 5A). The mean thick-
ness of the EL from the same series of donors was 134.2
nm (SD 32.0 nm) in the macula and 391.7 nm (SD
120.1 nm) in the extramacula (Figure 5B). This represents
a statistically significant difference in both the integrity
(P0.00001) and thickness (P0.00001) of the EL
between the macular and extramacular regions. When
the unaffected donors were split into young and age-
matched groups, aging effects in both the macular and
extramacular regions were observed. The thickness, but
not the integrity, of the EL was significantly higher in the
age-matched group, as compared to the young group, in
both the macular and peripheral regions (P0.033 and
P0.001, respectively). Scatter plots comparing the mean
integrity and mean thickness values for each donor (Figure
6) suggest that a strong relationship exists between integrity
and thickness of the EL in all regions in which these param-
eters were measured (exponential fit R
2
0.8735).
EL Integrity and Thickness in the Macular
Regions of Donors with Early and Advanced
Forms of AMD
In the macular area, there was an apparent thinning of EL
thickness and a loss of EL integrity from elderly normal
individuals, through early-stage AMD to late-stage AMD
(Table 1; Figures 7, 8, 9, and 10). For macular integrity,
significant differences were observed between unaf-
fected, age-matched controls and the following: all AMD
(P0.003), early-stage AMD (P0.028), late-stage
Figure 3. Values for mean integrity (percent; A,B) and thickness (nm; C,D)
of the elastic layer derived from the 14-year-old donor described in Figure
1B. Plots Aand Cdepict measurements taken from the inferior ora serrata to
the superior ora serrata, passing through the fovea (0), whereas plots Band
Dshow measurements spanning between the nasal and temporal ora serrata,
passing through the fovea (0) and optic nerve head (brackets; 5mmto7
mm). Note the extremely low integrity of the EL in the fovea (Aand B;
spanning between 2mmand2 mm) and adjacent to the optic nerve head
on the nasal side (8mmto10 mm in B). A step increase in integrity
occurs in the vicinity of the major retinal vessels (⬃⫺6mmand6mminA
and B). The EL is thinnest in the macula area and increases abruptly in the
region of the vascular arcades (⬃⫺6mmto6mminCand D).
Figure 4. A: A derived, color-coded schematic of the integrity of the elastic layer
(relative percent scale shown to right) of Bruch’s membrane based on the data
shown in Figure 3, A and B. The black circle represents the optic nerve head
and the curved black lines emanating from it are the approximate positions of
the major retinal vessels. Note that the integrity is lowest in the foveal region
(arrow), central macula, and on the nasal aspect of the optic nerve head. These
regions of decreased integrity correspond spatially to the distribution of the
majority of macular lesions, such as GA (B;arrows depict the margin of macular
atrophy) and CNVMs (C;arrow) that occur in individuals with AMD.
Topographical Variation in Bruch’s Membrane 245
AJP January 2005, Vol. 166, No. 1
AMD (P0.003), active CNVM (P0.02), and CNV/DS
(P0.003). For macular thickness, significant differ-
ences were observed between age-matched controls
and donors with CNV/DS (P0.008), the latter which
accounted for the apparent significant differences ob-
served in the AMD (P0.035) and late-stage AMD (P
0.024) groups (Table 1). These changes in EL thickness
were in the opposite direction to those seen in normal
aging. In contrast to the macula, no significant differ-
ences were found in the extramacular region between
age-matched donors and those with early- or late-stage
AMD. Importantly, no significant differences in EL param-
eters were observed between unaffected, age-matched
controls and individuals with GA. Further analyses were
made between early-stage AMD and late-stage AMD,
between active CNVM and CNV/DS, between all CNV
and GA, between active CNVM and GA, and between
CNV/DS and GA (Table 1). No significant differences in
macular or extramacular EL thickness or integrity were
revealed in these comparisons.
EL Gap Length in the Macular and Peripheral
Regions of Donors with and without AMD
The largest gap length, or discontinuity, in the EL was
assessed for each donor used in this study (Figures 9
and 10). There were no significant differences in largest
elastin layer gap in the extramacular region between the
various control and affected groups. In the macular area,
the largest gaps in the elastin layer were significantly
larger in the early AMD (P0.0001), active CNV (P
0.0001), and CNV/DS (P0.0001) groups (in which the
average gap length was 9to10
m), as compared to
both the age-matched and young unaffected control
groups (in which the average gap width was 4 to 5
m).
No significant differences were observed between do-
nors in the unaffected, age-matched control group and
those in the classic GA group (P0.11).
Discussion
Bruch’s membrane likely provides a scaffold for RPE
adhesion, regulates the diffusion of molecules between
the choroid and retina, and serves as a physical barrier to
cell movement, restricting the passage of cells between
the choroid and retina.
40
Numerous changes have been
described at this interface that have the potential to dis-
rupt the normal physiology of Bruch’s membrane and its
surrounding tissues
3–13,16–19,21,22,25,28,41,42
, however
few of them help to explain the predilection of the macular
Bruch’s membrane to degradation and dysfunction that
occurs in individuals afflicted with AMD. To pursue pre-
liminary findings suggesting the presence of topograph-
ical variation in the EL between the macular and extra-
macular regions we rigorously examined the morphology
of this extracellular structure in a large series of human
donor eyes.
The Macular EL Is Thinner and More Porous in
All Individuals
In this study, we have found that the EL of Bruch’s mem-
brane is three to six times thinner and two to five times
more porous in the macular region than it is in the peripheral
region at all ages. This profound difference was not antici-
pated based on previous investigations.
4,14,17,27,43–47
It is
intriguing to speculate about the mechanisms that might
give rise to the observed topographical variation in the
Figure 5. Scatter plots showing values of EL integrity (top; percent) and thick-
ness (bottom; nm) derived from 121 individual donors, aged 1 day to 99 years.
Note that the integrity of the EL in the macular region is approximately three
times lower, on average, than that of the extramacular regions in all donors. The
thickness of the EL in the macular regions is approximately four to six times
lower, on average, than that of the extramacular region in all donors.
Figure 6. Scatter plot showing the relationship between EL integrity and
thickness for all donors examined in this investigation. Note that there is a
strong relationship between these two parameters in all regions measured
(exponential fit R
2
0.8735).
246 Chong et al
AJP January 2005, Vol. 166, No. 1
EL. One explanation is that the majority of the elastin in
Bruch’s membrane is synthesized at a time before that of
the final differentiation of the macula, which occurs dur-
ing the first few years of postnatal life.
48
This concept is
supported by the fact that the majority of elastin is de-
posited in most tissues and organs during the latter
stages of gestation.
49
Elastin gene reactivation and neo-
synthesis does occur postnatally, typically in response to
injury and inflammation, however its deposition rarely
results in the reformation of ordered elastin fibers.
50,51
It is also conceivable that differential thinning of
Bruch’s membrane-associated elastin occurs during
postnatal growth of the eye and this event is more pro-
nounced in the macula because of differential rates of
growth and/or stretching in this region. This process may
be exacerbated in myopia, a condition characterized by
an increased axial length of the eye and oftentimes by the
secondary formation of macular atrophy and CNVMs.
52
Our data also suggest that an interesting regional re-
lationship between the porosity and thickness of the EL
exists (Figure 6). Increasing integrity is associated with
increasing thickness in a nearly exponential manner from
the macula to the extramacular region. This relationship,
which might prove to be predictive of AMD, will be ex-
amined in more detail in future studies.
The Macular EL in Individuals with AMD
This study also shows that the integrity of the macular EL
is significantly lower in individuals with early-stage AMD,
as compared to age-matched controls, whereas both the
thickness and integrity of the EL are significantly lower in
individuals with late-stage AMD, including those with ac-
tive CNVMs and DSs (except for those with classic GA,
as discussed below). It will be valuable to understand
better the mechanisms that lead to even greater porosity
Table 1. Comparative Analyses of Elastic Lamina Parameters Using the Mann-Whitney U-Test
Macular
thickness
Macular
integrity
Peripheral
thickness
Peripheral
integrity
Aging comparisons
62 Young versus 62 age-matched 0.033 0.34 0.001 0.28
Age-matched to AMD comparisons
62 Age-matched versus all AMD 0.035 0.003 0.77 0.94
62 Age-matched versus early-stage AMD 0.19 0.028 0.42 0.49
62 Age-matched versus late-stage AMD 0.024 0.003 0.92 0.55
62 Age-matched versus GA 0.15 0.10 0.99 0.95
62 Age-matched versus active CNVM 0.44 0.02 0.42 0.94
62 Age-matched versus CNV/DS 0.008 0.003 0.79 0.24
AMD subgroup comparisons
Early-stage AMD versus late-stage AMD 0.25 0.45 0.40 0.12
Active CNVM versus CNV/DS 0.089 0.64 0.35 0.51
CNVM and CNV/DS versus GA 0.61 0.41 0.72 0.35
Active CNVM versus GA 0.59 0.64 0.46 0.92
CNV/DS versus GA 0.28 0.40 0.98 0.23
Significant differences are bolded.
Figure 7. Top: Box and whiskers plots showing data for EL thickness (A,B)
and integrity (C,D) derived from the macular (A,C) and extramacular (B,D)
regions of unaffected donors 62 years, age-matched donors 62 years,
donors with early-stage AMD, and donors with late-stage AMD (includes
donors with GA, active CNVMs, and DSs). Median values are shown as bold
red lines within each box), first and third quartile values are represented as
blue and magenta, respectively, within the solid box, the minimum and
maximum observation ranges are shown as whiskers and extreme values are
represented as solitary, horizontal lines. One young donor with an extreme
macular elastic layer thickness of 275 nm and one age-matched control donor
with an extreme macular elastic layer thickness of 409 nm were omitted from
graph Afor scaling purposes.
Figure 8. Bottom: Box and whiskers plots showing data for EL thickness (A,
B) and integrity (C,D) derived from the macular (A,C) and extramacular (B,
D) regions of donors with early-stage AMD and subgroups of donors with
late-stage AMD, including active CNVMs [CNV (act)], DSs ( CNV/DS), and GA.
Topographical Variation in Bruch’s Membrane 247
AJP January 2005, Vol. 166, No. 1
and thinning of the macular EL in individuals with AMD.
One explanation is that the overall integrity and thickness
of the macular EL do not decrease with advancing age in
individuals who develop AMD, but rather, that these pa-
rameters are lower in affected individuals from the time of
birth. This could occur, for example, if ocular elastin
synthesis was impaired during gestation, if specific iso-
forms of elastin were synthesized that predispose indi-
viduals to disease, or if one or more components of the
elastin synthetic pathway were dysfunctional because of
mutations in the genes encoding elastin or elastin-asso-
ciated proteins. It has been established experimentally,
for example, that exposure to increased amounts of vita-
min D during gestation results in a reduction of aortic
elastin content.
53
Similarly, it has been proposed that the
synthesis of aortic elastin is deficient in fetuses with im-
paired growth.
54
Moreover, the expression of tissue-spe-
cific isoforms of tropoelastin,
55
or the enhanced expres-
sion of the wrong isoform might predispose one to AMD.
Yet another explanation for the observed decrease in
macular EL integrity in individuals with AMD is that the
elastin layer is degraded in these patients more rapidly
than it is in unaffected individuals. Along these lines,
we
20,37
and others
47,56– 60
have provided evidence that
inflammatory and immune-mediated processes, particu-
larly complement activation,
30,61
are associated with the
development of AMD. These processes are prominent at
the RPE-Bruch’s membrane interface, and are typically
more robust in the macular region than in the extramacu-
lar region. A potential consequence of these chronic
inflammatory processes, if analogous to those that occur
in a variety other age-related disease processes,
62,63
would be an overall disruption, degradation, and/or re-
modeling of Bruch’s membrane-associated elastin and
collagen. In cutus laxa, for example, a robust and dra-
matic loss of cutaneous elastic fibers is caused by local
inflammation.
64
If local, chronic inflammation participates in the de-
struction of the Bruch’s membrane, one would predict
that macular CNVM formation would be associated with
other chronic, systemic inflammatory diseases. Indeed,
the development of neovascular membranes is not lim-
ited to AMD.
65,66
On review, there are numerous immune-
mediated and inflammation-based diseases including
presumed ocular histoplasmosis syndrome, membrano-
proliferative glomerulonephritis, rubella, toxoplasma reti-
nochoroiditis, sarcoidosis, Vogt-Koyanagi-Harada dis-
ease, birdshot choroidopathy, Bechet’s disease, chronic
uveitis, and bacterial endocarditis,
67–69
in which CNVMs
can form. Importantly, the majority of these CNVMs form
within the macula, an issue that is addressed in more
detail below.
The Macular Elastic Layer in Classic GA
These data also show that the integrity and thickness of the
macular EL in individuals with GA does not vary significantly
from those of unaffected, age-matched controls. These
data imply that differences or changes in the EL of individ-
uals with AMD may be restricted to a specific phenotype(s)
or genotype(s) of the disease. Importantly, they may also
help to explain the distinct pattern of macular degeneration
that is observed in individuals afflicted with GA.
Consequences of Decreased Macular EL
Integrity in AMD
Whatever the derivation of the unique macula-associated
features of Bruch’s membrane described herein, one
would predict that any degeneration-inducing processes
and/or modifications of the structural components of
Bruch’s membrane (eg, collagen and elastin), whether
genetic or acquired, might preferentially affect the overall
function of the macular region. One consequence of an
EL that is more porous in the macula might be that there
is less substrate for RPE cell adhesion. If proven to be the
case, this would help to explain the propensity for pig-
ment epithelial cell detachments, basal laminar deposit
accumulation, and soft drusen formation—all strong risk
factors for the development of AMD—to form within the
macula.
7,16
Compromised adhesion of macular RPE cells
might also help to explain the dysfunction of these cells
that occurs in AMD.
Figure 9. Transmission electron micrographs depicting the EL of Bruch’s mem-
brane in eyes from a 82-year-old, age-matched control donor (A), an 84-year-old
donor with early AMD (B), and an 83-year-old AMD donor with active CNV (C).
The elastic layers are depicted with arrows and longest gaps in macular EL
integrity (maximum gap lengths) for each donor are bracketed.
Figure 10. Bar graph depicting the average values for the largest gap length,
or discontinuity, in the EL of donors with early-stage AMD and subgroups of
donors with late-stage AMD (active CNVM, CNVM/DS, and GA). Significant
differences in maximum gap length are noted between the both control
groups (young and age-matched) and the early AMD, active CNVM, and
CNVM/DS groups (all P0.0001), but not between the age-matched control
group and the GA group (P0.11).
248 Chong et al
AJP January 2005, Vol. 166, No. 1
Another consequence of a more porous macular EL
might be the predilection of CNVMs to form within the
macula. CNV—a pathological process in which neoves-
sels emanate from the choroidal vasculature, breach the
barrier imposed by Bruch’s membrane, and grow into the
sub-RPE and subretinal spaces—is associated with a
variety of disease processes,
65,66
including AMD. CNVM
formation can occur secondary to genetic alteration and
degeneration of Bruch’s membrane, such as occur in
pseudoxanthoma elasticum, Ehlers Danlos syndrome,
Sorsby’s fundus dystrophy, and Menke’s disease,
70–74
suggesting that genetic conditions resulting in elastin
breakdown can predispose one to CNVM formation.
CNVM formation may also be acquired in certain situa-
tions. For example, disruption of Bruch’s membrane that
occurs as a consequence of mechanical stress in cho-
roidal rupture, myopia, cryoinjury, ocular massage, and
manipulation of the eye during cataract extraction is
often associated with the subsequent formation of
CNVM.
23,52,75,76
Similarly, CNVM formation can be in-
duced iatrogenically in human eyes
77
and experimentally
in monkey eyes by damage to Bruch’s membrane after
laser photocoagulation.
32
One recent study shows that
there is a predilection of the macula to a higher incidence
of CNVM formation after intense laser photocoagulation
in monkeys.
78
Moreover, in mouse models of CNV
79,80
the majority of data provide evidence that neither choroidal
nor retinal neovessels are capable of breaching Bruch’s
membrane unless it is first disrupted,
79–81
consistent with
the notion that a robust Bruch’s membrane may safeguard
against CNV, although one investigator has suggested that
mechanical disruption is not prerequisite.
82
Thus, it seems clear that the structural components of
Bruch’s membrane must be compromised for CNV to
occur and it is reasonable to postulate that a neovascular
breach of Bruch’s membrane is primarily opportunistic,
after a failure of its barrier function. However, a satisfac-
tory explanation for this topographical predilection of
CNVM formation to occur in the macula has not been
made previously.
We propose that the data related to macular EL integ-
rity may help to explain this predilection. In the proposed
model of inflammation-induced destruction of Bruch’s
membrane, as an example, the macular region might
likely be compromised before that of the extramacular
region, based on the morphological characteristics of
Bruch’s membrane described herein. In other words,
pan-choroidal inflammation would be expected to have a
more rapid and dramatic effect in the macula, as com-
pared to other regions, because of the more porous
nature of its elastin layer (Figure 11A). Ironically, the
elastin peptides that might be generated as a conse-
quence of elastin fiber destruction in the macula could
play a key role in subsequent neovascular events be-
cause of their highly angiogenic properties.
83,84
In addition, the observation that the largest gap
lengths (discontinuities) within the macular EL are signif-
icantly longer in individuals with AMD (with the exception
of eyes with GA) than in age-matched controls may also
help to explain the predilection toward CNVM formation in
some eyes. For a neovessel to grow from the choroid into
the sub-RPE space, its cells must be able to pass phys-
ically through Bruch’s membrane. Although the smallest
theoretical pore through which choroidal neovessel-as-
sociated endothelial cells can migrate has not been es-
tablished to our knowledge, it is intuitive that the larger
gaps observed in some AMD donors would more readily
allow penetration by developing neovascular fronds.
Along these lines, it is interesting to note that the majority
of images in the literature that depict early choroidal
neovessels penetrating Bruch’s membrane show gaps
greater than 5
m in diameter. Our data show that many
donors with AMD, except for those with GA, have EL gap
widths greater than 5
m. These data also imply that the
decreased integrity observed in many of our AMD donors
can be accounted for pores that are larger in diameter
than in the unaffected controls.
Summary
In summary, the results of this study have documented
the existence of unique structural features associated
with the macular EL of Bruch’s membrane. These at-
tributes correspond spatially to the distribution of the
majority of macular lesions, including GA, CNV, and DS
formation that are associated with AMD, a leading cause
of blindness throughout the world. Our current working
hypothesis is that insults to the structural integrity of
Bruch’s membrane, whether mechanical, inflammatory,
or compositional in nature, are prerequisite for invasion of
choroidal neovessels into the sub-RPE and/or subretinal
spaces, even in conditions in which blood vessel growth
is abnormally enhanced. By extension, we suggest that
once the macular EL is compromised, CNVM formation is
likely to reoccur after mechanical or pharmaceutical de-
struction of an initial neovascular membrane (Figure
Figure 11. Models depicting possible scenarios that might lead to the en-
hanced susceptibility of the macula to CNVM formation (A) and CNVM
recurrence (B). We have proposed that AMD-associated pathways, such as
inflammation, that degrade or disrupt elastin might affect the barrier function
of Bruch’s membrane in the macular region before that of the extramacular
region because the elastic layer is the thinnest and most porous in this region
(Aand B,lane 1). The resulting breaches of Bruch’s membrane in the
macula (asterisks;Aand B,lane 2) might allow CNVMs to penetrate
Bruch’s membrane and to grow into the subretinal and/or sub-RPE spaces. In
treatments intended to destroy these neovessels, such as laser photocoagu-
lation and photodynamic therapy (wavy lines;B,lane 3), the breached
region of Bruch’s membrane might be replaced by fibrotic scar tissue (hatch-
es;B,lane 4). Subsequently, the process of elastin degradation and neovas-
cularization would continue at thinned, porous regions of the EL lying
adjacent to that of the original vascular penetration (B,lanes 4 and 5).
Ultimately, this process might continue until a scar formed over that portion
of the macula (hatches;B,lane 6) where the EL was initially the thinnest
and most porous.
Topographical Variation in Bruch’s Membrane 249
AJP January 2005, Vol. 166, No. 1
11B). Indeed, this phenomenon of macular neovessel
recurrence is frequently observed after CNVM treatment
modalities.
85,86
We conclude that the quantitatively dis-
tinct features of the EL may help to explain the enhanced
susceptibility of the macula to degeneration in AMD and
other diseases characterized by neovascularization in
the macular region.
Acknowledgments
We thank Dr. Stephen Russell for stimulating discussions
and for evaluation of human donor eyes; Dr. Robert
Mecham for his kind gift of anti-elastin polyclonal anti-
bodies; James Borchardt, Ryan Lee, Amber Bain, Sheri
McCormick, and Aleks Rozek for logistical assistance;
Sepidah Amin and Karen Gehrs for helpful discussions;
the staffs of the Iowa Lions Eye Bank, MidAmerica Trans-
plant Services, the Heartland Eye Bank, the Central Flor-
ida Lions Eye and Tissue Bank, and the Virginia Eye Bank
for their dedication in procuring the eyes used in these
studies; and all of the families who unselfishly donated
eyes of loved ones to this study.
References
1. Marshall J, Hussain A, Starita C, Moore D, Patmore A: Aging and
Bruch’s membrane. The Retinal Pigment Epithelium. Edited by M
Marmor, T Wolfensberger. New York, Oxford University Press, 1988,
pp 669 – 692
2. Guymer R, Bird A: Bruch’s membrane, drusen, and age-related mac-
ular degeneration. The Retinal Pigment Epithelium. Edited by M Mar-
mor, T Wolfensberger. New York, Oxford University Press, 1988, pp
693–705
3. Hogan M: Bruch’s membrane and disease of the macula. Trans
Ophthalmol Soc UK 1967, 87:113–161
4. Hogan M, Alvarado J: Studies on the human macula. Aging changes
in Bruch’s membrane. Arch Ophthalmol 1967, 77:410 420
5. Guymer R, Luthert P, Bird A: Changes in Bruch’s membrane and
related structures with age. Prog Retinal Eye Res 1999 18:59 –90
6. Sarks S: Ageing and degeneration in the macular region: a clinico-
pathological study. Br J Ophthalmol 1976, 60:324 –341
7. Green W, McDonnell P, Yeo J: Pathologic features of senile macular
degeneration. Ophthalmology 1985, 92:615– 627
8. Grindle C, Marshall J: Ageing changes in Bruch’s membrane and their
functional implications. Trans Ophthalmol Soc UK 1978, 98:172–175
9. Feeney-Burns L, Ellersieck M: Age-related changes in the ultrastruc-
ture of Bruch’s membrane. Am J Ophthalmol 1985, 100:686 697
10. Pauleikhoff D, Sheraidah G, Marshall J, Bird A, Wessing A: Biochem-
ical and histochemical analysis of age related lipid deposits in
Bruch’s membrane. Ophthalmologe 1994, 91:730 –734
11. Ramrattan R, van der Schaft T, Mooy C, de Bruijn W, Mulder P, de
Jong P: Morphometric analysis of Bruch’s membrane, the choriocap-
illaris, and the choroid in aging. Invest Ophthalmol Vis Sci 1994,
35:2857–2864
12. Karwatowski W, Jeffries T, Duance V, Albon J, Bailey A, Easty D: Prep-
aration of Bruch’s membrane and analysis of the age-related changes in
the structural collagens. Br J Ophthalmol 1995, 79:944 –952
13. Moore D, Hussain A, Marshall J: Age-related variation in the hydraulic
conductivity of Bruch’s membrane. Invest Ophthalmol Vis Sci 1995,
36:1290 –1297
14. Kliffen M, van der Schaft TL, Mooy CM, de Jong PT: Morphologic
changes in age-related maculopathy. Microsc Res Tech 1997, 36:
106 –122
15. Kamei M, Apte SS, Rayborn ME, Lewis H, Hollyfield JG: TIMP-3
accumulation in Bruch’s membrane and drusen in eyes from normal
and age-related macular degeneration donors. Degenerative Retinal
Diseases. Edited by MM LaVail, JG Hollyfield, RE Anderson. New
York, Plenum Press, 1997, pp 11–15
16. Zarbin M: Age related macular degeneration: a review of pathogen-
esis. Eur J Ophthalmol 1998, 8:199 –206
17. Spraul C, Lang G, Grossniklaus H, Lang G: Histologic and morpho-
metric analysis of the choroid, Bruch’s membrane, and retinal pig-
ment epithelium in postmortem eyes with age-related macular degen-
eration and histologic examination of surgically excised choroidal
neovascular membranes. Surv Ophthalmol 1999, 44:S10 –S32
18. Okubo A, Rosa R, Bunce C, Alexander R, Fan J, Bird A, Luthert P:
Relationship of age changes in retinal pigment epithelium and
Bruch’s membrane. IOVS 1999, 40:443– 449
19. Curcio C, Millican C, Bailey T, Kruth H: Accumulation of cholesterol
with age in human Bruch’s membrane. Invest Ophthalmol Vis Sci
2001, 42:265–274
20. Hageman G, Luthert P, Chong N, Johnson L, Anderson D, Mullins R:
An integrated hypothesis that considers drusen as biomarkers of
immune-mediated processes at the RPE-Bruch’s membrane interface
in aging and age-related macular degeneration. Prog Ret Eye Res
2001, 20:705–732
21. Hussain A, Rowe L, Marshall J: Age-related alterations in the diffu-
sional transport of amino acids across the human Bruch’s-choroid
complex. J Opt Soc Am 2002, 19:166 –172
22. Handa J, Verzijl N, Matsunaga H, Aotaki-Keen A, Lutty G, te Koppele
J, Miyata T, Hjelmeland L: Increase in the advanced glycation end
product pentosidine in Bruch’s membrane with age. Invest Ophthal-
mol Vis Sci 1999, 40:775–779
23. Yamamoto T, Yamashita H: Pseudopodia of choriocapillary endothe-
lium. Jpn J Ophthalmol 1989, 33:327–336
24. Sarks J, Sarks S, Killingsworth M: Evolution of geographic atrophy of
the retinal pigment epithelium. Eye 1988, 2:552–577
25. Green W, Enger C: Age-related macular degeneration histopatho-
logic studies. Ophthalmology 1993, 100:1519 –1535
26. Bressler N, Silva J, Bressler S, Fine S, Green W: Clinicopathologic
correlation of drusen and retinal pigment epithelial abnormalities in
age-related macular degeneration. Retina 1994, 14:130 –142
27. Spraul C, Grossniklaus H: Characteristics of drusen and Bruch’s
membrane in postmortem eyes with age-related macular degenera-
tion. Arch Ophthalmol 1997, 115:267–273
28. Curcio C, Millican C: Basal linear deposit and large drusen are
specific for early age-related maculopathy. Arch Ophthalmol 1999,
117:329 –339
29. Hageman G, Mullins R, Russell S, Johnson L, Anderson D: Vitronectin
is a constituent of ocular drusen and the vitronectin gene is ex-
pressed in human retinal pigmented epithelial cells. FASEB J 1999,
13:477– 484
30. Mullins R, Anderson D, Russell S, Hageman G: Ocular drusen contain
proteins common to extracellular deposits associated with athero-
sclerosis, elastosis, amyloidosis, and dense deposit disease. FASEB
J 2000, 14:835– 846
31. Bhagat N, Flaxel C: Nonexudative macular degeneration. Age-Re-
lated Macular Degeneration. Edited by J Lim. New York, Marcel
Dekker, 2002, pp 67– 82
32. Ryan S: Subretinal neovascularization. Natural history of an experi-
mental model. Arch Ophthalmol 1982, 100:1804 –1809
33. Malthieu D, Turut P, Francois P: Subretinal neovessels caused by
photocoagulation. Bull Soc Ophthalmol Fr 1988, 88:645– 646
34. Lee S, Shen W, Yeo I, Lai C, Mathur R, Tan D, Constable I, Rakoczy
P: Predilection of the macular region to high incidence of choroidal
neovascularization after intense laser photocoagulation in the mon-
key. Invest. Ophthalmol Vis Sci 2003, 44:E-abstract 3945
35. Smith W, Assink J, Klein R, Mitchell P, Klaver C, Klein B, Hofman A,
Jensen S, Wang J, de Jong P: Risk factors for age-related macular
degeneration: pooled findings from three continents. Ophthalmology
2001, 108:697–704
36. Bird A, Bressler N, Bressler S, Chisholm I, Coscas G, Davis M, de
Jong P, Klaver C, Klein B, Klein R, Mitchell P, Sarks J, Sarks S,
Soubrane G, Taylor H, Vingerling J: An international classification and
grading system for age-related maculopathy and age-related macu-
lar degeneration. The International ARM Epidemiological Study
Group. Surv Ophthalmol 1995, 39:367–374
37. Anderson D, Mullins R, Hageman G, Johnson LV: A role for local
inflammation in the formation of drusen in the aging eye. Am J
Ophthalmol 2002, 134:411– 431
38. Hageman G, Mullins R: Molecular composition of drusen as related to
substructural phenotype. Mol Vis 1999, 5:28
250 Chong et al
AJP January 2005, Vol. 166, No. 1
39. Russell S, Mullins R, Schneider B, Hageman G: Basal laminar drusen
are indistinguishable in location, substructure, and composition from
drusen associated with aging and age-related macular degeneration.
Am J Ophthalmol 2002, 129:205–214
40. Goldberg MF: Bruch’s membrane and vascular growth. Invest Oph-
thalmol 1976, 15:443– 446
41. Yamomoto T, Yamashita H: Scanning electron microscopic observa-
tion of Bruch’s membrane with the osmium tetroxide treatment. Br J
Ophthalmol 1989 73:162–167
42. Kamei M, Hollyfield J: TIMP-3 in Bruch’s membrane: changes during
aging and in age-related macular degeneration. Invest Ophthalmol
Vis Sci 1999, 40:2367–2375
43. Tarkkanen A, Vannas S: Ultrastructure of Bruch’s membrane in senile
macular degeneration. Acta Ophthalmol 1967, 45:694 698
44. Hollenberg M, Burt W: The fine structure of Bruch’s membrane in the
human eye. Can J Ophthalmol 1969, 4:296 –306
45. Bonnet M, Schiffer H: Modifications du tissu elastique de la mem-
brane de Bruch dans la region maculaire en fonction de l’age. Bull
Mem Soc Fr Ophthalmol 1973, 86:353–366
46. Newsome D, Huh W, Green W: Bruch’s membrane age-related
changes vary by region. Curr Eye Res 1987, 6:1211–1221
47. Killingsworth M, Sarks J, Sarks S: Macrophages related to Bruch’s
membrane in age-related macular degeneration. Eye 1990, 4:613– 621
48. Hendrickson A, Yuodelis C: The morphological development of the
human fovea. Ophthalmology 1984, 91:603– 612
49. Davidson JM, Quaglino JD, Giro MG: Elastin repair. Wound Healing:
Biochemical and Clinical Aspects. Edited by IK Cohen, RE Di-
egelman, WJ Lindblad. Philadelphia, W.B. Saunders, 1992
50. Quaglino Jr D, Nanney LB, Kennedy R, Davidson JM: Transforming
growth factor-beta stimulates wound healing and modulates extracel-
lular matrix gene expression in pig skin. I. Excisional wound model.
Lab Invest 1990, 63:307–319
51. Pasquali Ronchetti I, Quaglino Jr D, Dyne KM, Zanaboni G, Cetta G:
Ultrastructural studies on dermis from prolidase deficient subjects. J
Submicrosc Cytol Pathol 1991, 23:439 445
52. Yoshida T, Ohno-Matsui K, Ohtake Y, Takashima T, Futagami S, Baba
T, Yasuzumi K, Tokoro T, Mochizuki M: Long-term visual prognosis of
choroidal neovascularization in high myopia: a comparison between
age groups. Ophthalmology 2002, 109:712–719
53. Norman P, Moss I, Sian M, Gosling M, Powell J: Maternal and post-
natal vitamin D ingestion influences rat aortic structure, function and
elastin content. Cardiovasc Res 2002, 55:369 –374
54. Martyn C, Greenwald S: Impaired synthesis of elastin in walls of aorta
and large conduit arteries during early development as an initiating
event in pathogenesis of systemic hypertension. Lancet 1997, 350:
953–955
55. Parks W, Roby J, Wu L, Grosso L: Cellular expression of tropoelastin
mRNA splice variants. Matrix 1992, 55:156 –162
56. Penfold P, Madigan M, Gillies M, Provis J: Immunological and aetio-
logical aspects of macular degeneration. Prog Ret Eye Res 2001,
20:385– 414
57. Heriot W, Henkind P, Bellhorn R, Burns M: Choroidal neovasculariza-
tion can digest Bruch’s membrane. A prior break is not essential.
Ophthalmology 1984 91:1603–1608
58. Penfold P, Killingsworth M, Sarks S: An ultrastructural study of the role
of leucocytes and fibroblasts in the breakdown of Bruch’s membrane.
Aust J Ophthalmol 1984, 12:23–31
59. Penfold P, Killingsworth M, Sarks S: Senile macular degeneration. The
involvement of giant cells in atrophy of the retinal pigment epithelium.
Invest Ophthalmol Vis Sci 1986, 27:364 –371
60. Dastgheib K, Green W: Granulomatous reaction to Bruch’s mem-
brane in age-related macular degeneration. Arch Ophthalmol 1994,
112:813– 818
61. Johnson L, Ozaki S, Staples M, Erickson P, Anderson D: A potential
role for immune complex pathogenesis in drusen formation. Exp Eye
Res 2000, 70:441– 449
62. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole G, Cooper N,
Eikelenboom P, Emmerling M, Fiebich B, Finch C, Frautschy S, Griffin
W, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie I,
McGeer P, O’Banion M, Pachter J, Pasinetti G, Plata-Salaman C,
Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van
Muiswinkel F, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk
G, Wyss-Coray T: Inflammation and Alzheimer’s disease. Neurobiol
Aging 2000, 21:383– 421
63. Phipps R: Atherosclerosis: the emerging role of inflammation and the
CD40-CD40 ligand system. Proc Natl Acad Sci USA 2000, 97:6930
6932
64. Uitto J, Fazio M, Christiano A: Cutis laxa and premature aging syn-
dromes. Connective Tissue and Its Heritable Disorders: Molecular,
Genetic, and Medical Aspects. Edited by P Royce, B Steinmann. New
York, Wiley-Liss, 1993, pp 409 423
65. Green W, Wilson D: Choroidal neovascularization. Ophthalmology
1986, 93:1169 –1176
66. Dolan B: Choroidal neovascularization not associated with age-re-
lated macular degeneration. Optom Clin 1996, 5:55–76
67. Meredith T, Green W, Key S, Dolin G, Maumenee A: Ocular his-
toplasmosis: clinicopathologic correlation of 3 cases. Surv Oph-
thalmol 1977, 22:189 –205
68. Olk R, Burgess D, McCormick P: Subfoveal and juxtafoveal subretinal
neovascularization in the presumed ocular histoplasmosis syndrome.
Visual prognosis. Ophthalmology 1984, 91:1592–1602
69. Ryan SJ: The Retina. St. Louis, C.V. Mosby, 1989
70. Erkkila H, Raitta C, Niemi K: Ocular findings in four siblings with
pseudoxanthoma elasticum. Acta Ophthalmol (Copenh) 1983, 61:
589 –599
71. Weber B, Vogt G, Pruett R, Stohr H, Felbor U: Mutations in the tissue
inhibitor of metalloproteinases-3 (TIMP3) in patients with Sorsby’s
fundus dystrophy. Nat Genet 1994, 8:352–356
72. Chong N, Alexander R, Gin T, Bird A, Luthert P: TIMP-3, collagen, and
elastin immunohistochemistry and histopathology of Sorsby’s fundus
dystrophy. Invest Ophthalmol Vis Sci 2000, 41:898 –902
73. Wong S, Fong K, Lee N, Gregory-Evans K, Gregory-Evans C: Suc-
cessful photodynamic therapy for subretinal neovascularization due
to Sorsby’s fundus dystrophy: 1 year follow up. Br J Ophthalmol 2003,
87:796 –797
74. Wray S, Kuwabara T, Sanderson P: Menkes’ kinky hair disease: a light
and electron microscopic study of the eye. Invest Ophthalmol 1976,
15:128 –138
75. Cohen S: Etiology of choroidal neovascularization in young patients.
Ophthalmology 1996, 103:1241–1244
76. Klein R, Klein B, Wong T, Tomany S, Cruickshanks K: The association
of cataract and cataract surgery with the long-term incidence of
age-related maculopathy: the Beaver Dam eye study. Arch Ophthal-
mol 2002, 120:1551–1558
77. Lim J: Iatrogenic choroidal neovascularization. Surv Ophthalmol
1999, 44:95–111
78. Shen W, Lee S, Yeo I, Lai C, Mathur R, Tan D, Constable I, Rakoczy
P: Predilection of the macular region to high incidence of choroidal
neovascularization after intense laser photocoagulation in the mon-
key. Arch Ophthalmol 2004, 122:353–360
79. Schwesinger C, Yee C, Rohan R: Intrachoroidal neovascularization in
transgenic mice overexpressing vascular endothelial growth factor in
the retinal pigment epithelium. Am J Pathol 2001, 158:1161–1172
80. Ohno-Matsui K, Hirose A, Yamamoto S, Saikia J, Okamoto N, Gehl-
bach P, Duh E, Hacket S, Chang M, Bok D, Zack D, Campochiaro P:
Inducible expression of vascular endothelial growth factor in adult
mice causes severe proliferative vitreoretinopathy and retinal detach-
ment. Am J Pathol 2002, 160:711–719
81. Kwak N, Okamoto N, Wood J, Campochiaro P: VEGF is major stim-
ulator in model of choroidal neovascularization. Invest Ophthalmol Vis
Sci 2000, 41:3158 –3164
82. Spilsbury K, Garrett K, Shen W, Constable I, Rakoczy P: Overexpres-
sion of vascular endothelial growth factor (VEGF) in the retinal pig-
ment epithelium leads to the development of choroidal neovascular-
ization. Am J Pathol 2000, 157:135–144
83. Nackman G, Karkowski F, Halpern V, Gaetz H, Tilson M: Elastin
degradation products induce adventitial angiogenesis in the Anidjar/
Dobrin rat aneurysm model. Surgery 1997, 122:39 – 44
84. Tummalapalli C, Tyagi S: Responses of vascular smooth muscle cell
to extracellular matrix degradation. J Cell Biochem 1999, 75:515–527
85. Pollack A, Korte G, Weitzner A, Henkind P: Ultrastructure of Bruch’s
membrane after krypton laser photocoagulation. I. Breakdown of
Bruch’s membrane. Arch Ophthalmol 1986, 104:1372–1376
86. Lafaut B, Aisenbrey S, Vanden Broecke C, Di Tizio F, Bartz-Schmidt
K: Clinicopathologic correlation in exudative age-related macular
degeneration: recurrent choroidal neovascularization. Graefes Arch
Clin Exp Ophthalmol 2001, 239:5–11
Topographical Variation in Bruch’s Membrane 251
AJP January 2005, Vol. 166, No. 1
... Na/K adenosine triphosphatase (ATPase) pumps were found to be 40 to 60% more expressed in peripheral RPE regions, reflecting regional differences in ionic regulation (12,13). The elastic lamina of the Bruch's membrane was demonstrated to be three to six times thinner in the macula in comparison to the peripheral RPE, suggesting different molecular composition of the Bruch's membrane at different retinal locations (14,15). Consistently, elastin (ELN)-and collagen-related genes have been shown to be more highly expressed in peripheral RPE (15). ...
... To determine if the isolated RPE subpopulations also display molecular differences, we stained non-AMD RPE flatmounts for ELN and myocilin (MYOC) proteins, previously shown to have higher expression in midperipheral RPE regions compared to the macula (14,15,18). Antibodies against ELN and MYOC were validated on cryosections of non-AMD eyes (SI Appendix, Fig. S1 A and B). ...
... The presence of RPE subpopulations of different cell size suggests that these subpopulations are physiologically different. Evidence of regional RPE heterogeneity between central and peripheral areas of the monolayer has been described previously by gene expression (15, 17, 18, 48), lysosomal enzyme activity (11,49,50), Na/K ATPases expression (12), and Bruch's membrane thickness (14). In this study, we propose that different RPE subpopulations are differentially sensitive to different retinal diseases. ...
Article
Full-text available
Significance Retinal degenerative diseases affect specific regions of the retinal pigment epithelium (RPE), suggesting the presence of functionally different RPE subpopulations. To identify these subpopulations in human eyes, we generated the first complete morphometric map of the RPE at single-cell resolution using artificial intelligence–based software. We identified five concentric RPE subpopulations, including a ring of RPE cells with cell area similar to macula in the periphery of the eye. Moreover, we found that specific RPE subpopulations are differentially susceptible to monogenic and polygenic retinal diseases. The results obtained here will allow study of molecular and functional RPE differences responsible for regional retinal diseases and will help develop precise cell and gene therapies for specific degenerative eye diseases.
... CNV, it causes 90% of severe vision loss (8). ...
... Furthermore, it regulates molecule and substance diffusion between the RPE and choriocapillaris layer. The Bruch membrane inhibits cellular and vascular migration from the choroid into the subretinal or sub-RPE and vice versa (8,17). ...
... Age-related RPE and Bruch's membrane changes stimulate chronic low-grade inflammation or parainflammation that causes more damage to RPE cells, thereby creating a vicious cycle. This event amplifies and persists immune system activity against RPE and Bruch's membrane (8,21,22). RPE cells damage leads to drusenogenesis, a form of extracellular accumulation that lies between RPE and Bruch's membrane. ...
... It appears that the integrity of the Bruch membrane is essential for preventing the invasion of capillaries from the choroidal circulation into the retina. 9 The macula (also known as macula lutea) is a region of the retina measuring around 5.5 mm in diameter and corresponding to the major temporal arcades. The macula is qualified for the middle 15 to 20 degrees of the visual field and has the best resolution of the eye. ...
... The macula is qualified for the middle 15 to 20 degrees of the visual field and has the best resolution of the eye. 9 Histologically, it var-ies from the peripheral retina in that it contains multiple layers of retinal ganglion cells, which are neurons dedicated to transferring visual input to areas of the brain cortex. In addition, mature RPE cells in the peripheral retina, but not the central retina, are efficient in multiplying and migrating to the senescent areas in the central retina. ...
Article
Full-text available
One of the most complicated eye disorders is age-related macular degeneration (AMD) which is the leading cause of irremediable blindness all over the world in the elderly. AMD is classified as early stage to late stage (advanced AMD), in which this stage is divided into the exudative or neovascular form (wet AMD) and the nonexudative or atrophic form (dry AMD). Clinically, AMD primarily influences the central area of retina known as the macula. Importantly, the wet form is generally associated with more severe vision loss. AMD has a systemic component, where many factors, like aging, genetic, environment, autoimmune and non-autoimmune disorders are associated with this disease. Additionally, healthy lifestyles, regular exercise, maintaining a normal lipid profile and weight are crucial to decreasing the risk of AMD. Furthermore, therapeutic strategies for limiting AMD should encompass a variety of factors to avoid and improve drug interventions, and also need to take into account personalized genetic information. In conclusion, with the development of technology and research progress, visual impairment and legal blindness from AMD have been substantially reduced in incidence. This review article is focused on identifying and developing the knowledge about the association between genetics, and etiology with AMD. We hope that this review will encourage researchers and lecturers, open new discussions, and contribute to a better understanding of AMD that improves patients’ visual acuity, and upgrades the quality of life of AMD patients.
... It is not clear why such regional and cell type-preferred differences in PR loss occur in the face of generalized insults. Macular vulnerability could be related to specific structural and functional modifications in this area, such as differences in vascularization [8], Bruch's membrane thickness [9], increased light exposure [10], cone density [11], or increased phagocytic load because of the higher PR/retinal pigmented epithelium (RPE) ratio [12]. But it has not been generally considered that some intrinsic feature of cones could render them differentially sensitive to generalized insults. ...
... The retina, particularly the PR, contain the highest levels of DHA in the body [46,47,48]. This PUFA has been shown to improve neurological and ophthalmological development in children [46,47,48], and numerous animal studies have demonstrated beneficial effects upon PR degeneration [9,49,50,51]. DHA is a major source of neuroprotectin D1, which has been shown to enhance cell survival in a number of experimental models [52,53,54]. Furthermore, DHA is also a precursor for resolvins, another group of active substances which possess anti-inflammatory properties [55]. ...
Article
Full-text available
Purpose In many retinal pathological conditions, rod and cone degeneration differs. For example, the early-onset maculopathy Stargardts disease type 1 (STGD1) is typified by loss of cones while rods are often less affected. We wanted to examine whether there exist intrinsic membrane differences between rods and cones that might explain such features. Methods Abca4 mRNA and protein levels were quantified in rod- and cone-enriched samples from wild-type and Nrl−/− mice retinas; rod- and cone-enriched outer segments (ROS and COS respectively) were prepared from pig retinas, and total lipids were analyzed by flame ionization, chromatography, and tandem mass spectrometry. Immunohistochemical staining of cone-rich rodent Arvicanthis ansorgei retinas was conducted, and ultra-high performance liquid chromatography of lipid species in porcine ROS and COS was performed. Results Abca4 mRNA and Abca4 protein content was significantly higher (50–300%) in cone compared to rod-enriched samples. ROS and COS displayed dramatic differences in several lipids, including very long chain poly-unsaturated fatty acids (VLC-PUFAs), especially docosahexaenoic acid (DHA, 22:6n-3): ROS 20.6% DHA, COS 3.3% (p < 0.001). VLC-PUFAs (> 50 total carbons) were virtually absent from COS. COS were impoverished (> 6× less) in phosphatidylethanolamine compared to ROS. ELOVL4 (“ELOngation of Very Long chain fatty acids 4”) antibody labelled Arvicanthis cones only very weakly compared to rods. Finally, there were large amounts (905 a.u.) of the bisretinoid A2PE in ROS, whereas it was much lower (121 a.u., ~ 7.5-fold less) in COS fractions. In contrast, COS contained fivefold higher amounts of all-trans-retinal dimer (115 a.u. compared to 22 a.u. in rods). Conclusions Compared to rods, cones expressed higher levels of Abca4 mRNA and Abca4 protein, were highly impoverished in PUFA (especially DHA) and phosphatidylethanolamine, and contained significant amounts of all-trans-retinal dimer. Based on these and other data, we propose that in contrast to rods, cones are preferentially vulnerable to stress and may die through direct cellular toxicity in pathologies such as STGD1.
... Точная причина ВМД до конца не изучена, но было выявлено несколько факторов риска, включая возраст, генетику, курение, ожирение и семейный анамнез заболевания [12,13]. Кроме того, окислительный стресс и воспаление были вовлечены в развитие и прогрессирование ВМД. ...
Article
Full-text available
В данном статье были проанализированы более 36 статьей, мы с коллегами дали основные понятие по применению стромальных/ стволовых клеток при возрастной макулярной дегенерации. Возрастная макулярная дегенерация (ВМД) – это дегенеративное заболевание глаз, которое поражает миллионы людей во всем мире, приводя к значительной потере зрения и инвалидности. Современные методы лечения ВМД, такие как терапия противосудорожным фактором роста эндотелия (анти-VEGF), направлены на замедление прогрессирования заболевания, но не способны полностью восстановить зрение. Следовательно, растет интерес к изучению потенциала стволовых клеток для лечения ВМД. Стволовые клетки обладают уникальной способностью дифференцироваться в различные типы клеток, включая клетки сетчатки, что делает их привлекательным кандидатом для регенеративной медицины в области офтальмологии. Были исследованы различные источники стволовых клеток, включая эмбриональные стволовые клетки, индуцированные плюрипотентные стволовые клетки и взрослые стволовые клетки, полученные из различных тканей.
... Experimental elevation of intraocular pressure above the perfusion pressure of the choroid is associated with increased ocular temperature in both monkeys and humans, indicating that the normal choroidal circulation serves as a heat sink (Parver 1991). The potential consequences of loss of choroidal flow in age-related macular degeneration (AMD) (discussed below) and its impact on temperature at the photoreceptor-RPE-choroid interface have not been fully explored; however, it is notable that RPE cells grown at elevated temperature show evidence of altered synthesis of extracellular matrix molecules (Sekiyama et al. 2012), which is a key feature of AMD (Chong et al. 2005, Grossniklaus et al. 1992, Sura et al. 2020). The choroid also plays a critical role in accommodation in nonmammalian species by rapid shape changes (Nickla & Wallman 2010), in contrast to the subtle and age-related thickness changes in humans, discussed below. ...
Article
The choriocapillaris, a dense capillary network located at the posterior pole of the eye, is essential for supporting normal vision, supplying nutrients, and removing waste products from photoreceptor cells and the retinal pigment epithelium. The anatomical location, heterogeneity, and homeostatic interactions with surrounding cell types make the choroid complex to study both in vivo and in vitro. Recent advances in single-cell RNA sequencing, in vivo imaging, and in vitro cell modeling are vastly improving our knowledge of the choroid and its role in normal health and in age-related macular degeneration (AMD). Histologically, loss of endothelial cells (ECs) of the choriocapillaris occurs early in AMD concomitant with elevated formation of the membrane attack complex of complement. Advanced imaging has allowed us to visualize early choroidal blood flow changes in AMD in living patients, supporting histological findings of loss of choroidal ECs. Single-cell RNA sequencing is being used to characterize choroidal cell types transcriptionally and discover their altered patterns of gene expression in aging and disease. Advances in induced pluripotent stem cell protocols and 3D cultures will allow us to closely mimic the in vivo microenvironment of the choroid in vitro to better understand the mechanism leading to choriocapillaris loss in AMD.
... Elastin makes up ~4% of the dry retinal tissue weight and has been detected at the basement membrane of retinal arterial walls (Chen and Weiland, 2014;Chong et al., 2005;Sagaties and Raviola, 1989). It has been suggested that smooth muscle cells and endothelial cells of retinal vessel walls might be involved in retinal elastin synthesis as they stain positive for elastin (Chen and Weiland, 2014). ...
Article
The extracellular matrix (ECM) and its turnover play a crucial role in the pathogenesis of several inflammatory diseases, including age-related macular degeneration (AMD). Elastin, a critical protein component of the ECM, not only provides structural and mechanical support to tissues, but also mediates several intracellular and extracellular molecular signaling pathways. Abnormal turnover of elastin has pathological implications. In the eye elastin is a major structural component of Bruch's membrane (BrM), a critical ECM structure separating the retinal pigment epithelium (RPE) from the choriocapillaris. Reduced integrity of macular BrM elastin, increased serum levels of elastin-derived peptides (EDPs), and elevated elastin antibodies have been reported in AMD. Existing reports suggest that elastases, the elastin-degrading enzymes secreted by RPE, infiltrating macrophages or neutrophils could be involved in BrM elastin degradation, thus contributing to AMD pathogenesis. EDPs derived from elastin degradation can increase inflammatory and angiogenic responses in tissues, and the elastin antibodies are shown to play roles in immune cell activity and complement activation. This review summarizes our current understanding on the elastases/elastin fragments-mediated mechanisms of AMD pathogenesis.
... It is associated with an overactive complement system, and an increase in circulating antibodies against certain epitopes, including elastin has been reported. Aging and AMD is associated with a loss of the elastin layer of Bruch's membrane (BrM) (Chong et al., 2005), which can be mimicked in the smoke-induced ocular pathology (SIOP) model in mouse. Of relevance, we previously showed that immunization with elastin peptide oxidatively modified by cigarette smoke (ox-elastin), exacerbated structural and functional damage in SIOP (Annamalai et al., 2020). ...
... It is associated with an overactive complement system, and an increase in circulating antibodies against certain epitopes, including elastin has been reported. Aging and AMD is associated with a loss of the elastin layer of Bruch's membrane (BrM) (Chong et al., 2005), which can be mimicked in the smoke-induced ocular pathology (SIOP) model in mouse. Of relevance, we previously showed that immunization with elastin peptide oxidatively modified by cigarette smoke (ox-elastin), exacerbated structural and functional damage in SIOP (Annamalai et al., 2020). ...
... The EL degrades with age as a normal phenomenon. However, it shows frequently thinning and fragmentation in AMD [37] . The abnormalities of elastin metabolism are not only seen in BM, but also could be observed as a systemic phenomenon in AMD. ...
Article
Full-text available
Age-related macular degeneration (AMD) is a progressive retinal disease, which is the leading cause of blindness in western countries. There is an urgency to establish new therapeutic strategies that could prevent or delay the progression of AMD more efficiently. Until now, the pathogenesis of AMD has remained unclear, limiting the development of the novel therapy. Bruch's membrane (BM) goes through remarkable changes in AMD, playing a significant role during the disease course. The main aim of this review is to present the crucial processes that occur at the level of BM, with special consideration of the lipid accumulation and protein modifications. Besides, some therapies targeted at these molecules and the construction of BM in tissue engineering of retinal pigment epithelium (RPE) cells transplantation were listed. Hopefully, this review may provide a reference for researchers engaged in pathogenesis or management on AMD.
Article
Clinicopathologic studies of eyes lead to a better understanding of the nature of the ophthalmoscopic and fluorescein angiographic features of drusen. A study was conducted to provide clinicopathologic correlation of drusen and present a classification of drusen based on clinical and histopathologic features. The macular areas of three eyes from two patients were serially sectioned and studied, and features were depicted in a two-dimensional map and compared with clinical findings. All three tyes had large drusen (> 63 microns) with soft morphologic characteristics (poorly demarcated borders) documented on fundus photographs. In both eyes from one patient, these large drusen corresponded to areas of focal retinal pigment epithelium hypopigmentation overlying Bruch's membrane, which was diffusely thickened throughout the macula. Similar findings were noted in another eye from a second patient, and additionally, where there was relatively marked fluorescein staining of large drusen on the late phase of an angiogram, the pathologic correlation demonstrated detachment of the thickened inner aspect of Bruch's membrane from the remainder of Bruch's membrane. Focal hyperpigmentation corresponded to areas of hypertrophy of the retinal pigment epithelium and to clusters of pigmented cells in the subretinal space and outer nuclear area. Diffuse thickening of the inner aspect of Bruch's membrane is associated with retinal pigment epithelial hypopigmentation, focal atrophy, and soft (large) drusen formation.
Chapter
Prevalence of AMD increases with age. Nonexudative AMD is the most common form of AMD. Factors associated with AMD include increased age, heredity, photic injury, nutrition, toxic insults, and cardiovascular risk factors. High-risk characteristics of drusen for development of CNV include: soft drusen, large drusen, greater than five drusen, confluence, and focal hyperpigmentation. Disappearance of drusen can occur spontaneously or may follow laser to drusen (CNVPT and PTAMD). Disappearance of drusen may result in geographic atrophy. Monitoring visual acuity and visual symptoms for the progression of AMD is of utmost importance in applying timely treatment.
Chapter
Age-related macular degeneration (ARMD) is the leading cause of central vision loss in individuals over the age of 50 in the Western hemisphere.1 ARMD is characterized by the accumulation of debris (drusen) within Bruch’s membrane during the early stages of the disease, with the subsequent development of choroidal neovascularization or atrophy of the choriocapillaris and retinal pigment epithelium (RPE) at later stages. The latter events are thought to be causally involved in the death of macular photoreceptors. While the ophthalmological changes during the progression of the various forms of ARMD have been described, and end-stage histopathological descriptions of ARMD are available, little is known about the cellular mechanisms associated with the normal maintenance and turnover of Bruch’s membrane, the events that lead to drusen formation, or the cause of subretinal neovascularization and atrophy of the RPE and choriocapillaris.
Article
Background: Delineation of the morphologic aspects of age-related macular degeneration (ARMD) is helpful in correlation with the clinical features and may contribute to understanding the pathogenesis. Methods: All eyes on file in the Eye Pathology Laboratory, Wilmer Institute, with the known features of ARMD were analyzed according to age, sex, and race. Methods included routine sections in the macula, stepped-serial and serial sections, and electron microscopy and two-dimensional reconstruction in selected cases. Results: There were 760 eyes with ARMD from 450 patients. Only 5.3% of these patients were black. Nodular drusen were observed in 6.2% of eyes (n = 47), basal laminar deposits in 54.7%, basal linear deposits in 27.6%, neovascularization in 38.2%, and disciform scars in 40.8%. Three types of soft drusen were identified and were observed in 28.0%. The mean diameter of the disciform scar was 3.73 mm, and mean thickness was 0.27 mm. In disciform scars greater than 0.2 mm in thickness, only approximately 25% of the surface of the scar had some remaining photoreceptor cells. Serous or hemorrhagic detachments were observed in 10.4% of eyes. Retinal pigment epithelial (RPE) atrophy without disciform scars was observed in 24.6% of eyes. Areolar atrophy was the most common feature observed in eves of black patients. Conclusion: This comprehensive histopathologic evaluation provides the tabulation of the various morphologic features of ARMD. Basal laminar deposit and basal linear deposit, but not nodular drusen, are important positive associations with choroidal neovascularization, disciform scarring, and visual loss. Preservation of photoreceptor cells was seen only over disciform scars 0.2 mm thickness or less.
Article
A common detection and classification system is needed for epidemiologic studies of age-related maculopathy (ARM). Such a grading scheme for ARM is described in this paper. ARM is defined as a degenerative disorder in persons ≥50 years of age characterized on grading of color fundus transparencies by the presence of the following abnormalities in the macular area: soft drusen ≥63μm, hyperpigmentation and/or hypopigmentation of the retinal pigment epithelium (RPE), RPE and associated neurosensory detachment, (peri)retinal hemorrhages, geographic atrophy of the RPE, or (peri)retinal fibrous scarring in the absence of other retinal (vascular) disorders. Visual acuity is not used to define the presence of ARM. Early ARM is defined as the presence of drusen and RPE pigmentary abnormalities described above; late ARM is similar to age-related macular degeneration (AMD) and includes dry AMD (geographic atrophy of the RPE in the absence of neovascular AMD) or neovascular AMD (RPE detachment, hemorrhages, and/or scars as described above). Methods to take and grade fundus transparencies are described.
Article
The structural changes in Bruch's membrane and the pigment epithelium associated with increasing age may be of importance in understanding the morphogenesis of disciform lesions. This study includes the electron microscopic examination of 73 human retinae obtained from both enucleation and cadaver eyes and comprises specimens from each of nine decades, the youngest eye being 19 years and the oldest 90 years.