Untitled
Untitled
MACULAR
DEGENERATION
SECOND EDITION
AGE-RELATED MACULAR
DEGENERATION
EDITORS
ERIC P. JABLON, MD
Partner
Retina Consultants of Charleston
Charleston, South Carolina
KENNETH A. SHARPE, MD
Partner
Retina Consultants of Charleston
Charleston, South Carolina
Care has been taken to confirm the accuracy of the information presented and to describe generally
accepted practices. However, the authors, editors, and publisher are not responsible for errors or omissions
or for any consequences from application of the information in this book and make no warranty, expressed
or implied, with respect to the currency, completeness, or accuracy of the contents of the publication.
Application of the information in a particular situation remains the professional responsibility of the
practitioner.
The authors, editors, and publisher have exerted every effort to ensure that drug selection and dosage set
forth in this text are in accordance with current recommendations and practice at the time of publication.
However, in view of ongoing research, changes in government regulations, and the constant flow of
information relating to drug therapy and drug reactions, the reader is urged to check the package insert for
each drug for any change in indications and dosage and for added warnings and precautions. This is
particularly important when the recommended agent is a new or infrequently employed drug.
Some drugs and medical devices presented in the publication have Food and Drug Administration (FDA)
clearance for limited use in restricted research settings. It is the responsibility of the health care provider to
ascertain the FDA status of each drug or device planned for use in their clinical practice.
To purchase additional copies of this book, call our customer service department at (800) 638-3030 or fax
orders to (301) 223-2320. International customers should call (301) 223-2300.
Visit Lippincott Williams & Wilkins on the Internet: at LWW.com. Lippincott Williams & Wilkins customer
service representatives are available from 8:30 am to 6 pm, EST.
10 9 8 7 6 5 4 3 2 1
DEDICATION
Madhu Amara, MD
University Retina
Oak Forest, Illinois
Harit K. Bhatt, MD
Clinical Assistant Professor
Illinois Eye and Ear Infirmary
University of Illinois at Chicago
Chicago, Illinois
Attending Physician/Partner
University Retina and Macula Associates
Bedford Park, Illinois
Wissam Charafeddin, MD
Retina Fellow
Retina Department
Centro de Oftalmologia Barraquer
Barcelona, Spain
Lucienne C. Collet, MD
Admiravisión
Department of Ophthalmology
Barcelona, Spain
Borja F. Corcóstegui, MD
Professor
Department of Ophthalmology
Universitat Autonomous Barcelona
Barcelona, Spain
Michelle L. Crouse, MA
Clinical Trial Coordinator
Retina/Vitreous
Retina Consultants of Charleston
Charleston, South Carolina
Matías E. Czerwonko, MD
Advisor
Department of Quality and Patient Safety
Hospital Universitario Austral
Buenos Aires, Spain
Diana V. Do, MD
Associate Professor of Ophthalmology
Wilmer Eye Institute
School of Medicine
Johns Hopkins University
Baltimore, Maryland
Jay S. Duker, MD
Professor
Department of Ophthalmology
School of Medicine
Tufts University
Chairman
Department of Ophthalmology
Tufts Medical Center
Boston, Massachusetts
Carlos F. Fernández, MD
Chief
Department of Ophthalmology
Clinica Oftalmolaser
Lima, Peru
Reinaldo A. Garcia, MD
Associate Professor
Retina and Vitreous Disease
Clinica Oftalmologica El Vinedo
Valencia, Edo. Carabobo, Venezuela
Gerardo Garcia-Aguirre, MD
Attending Physician
Retina Department
Asociacion para Evitar la Ceguera en Mexico
Clinical Professor of Ophthalmology
Escuela de Medicina
Instituto Tecnológico y de Estudios Superiores de Monterrey
Mexico City, Mexico
Jose M. Garcia-Gonzalez, MD
Retina Fellow
Section of Ophthalmology
University of Chicago
Chicago, Illinois
Sundeep Grandhe, MD
University Retina
Oak Forest, Illinois
Gabriela E. Granella, MD
Director and Retina Specialist
Oftalmológico Santa Lucía
Buenos Aires, Argentina
Craig M. Greven, MD
Professor and Chairman
Department of Ophthalmology
School of Medicine
Wake Forest University
Winston-Salem, North Carolina
Mariana Ingolotti, MD
Assistant Professor
Department of Pathophysiology
Austral University
Buenos Aires, Argentina
Eric P. Jablon, MD
Partner
Retina Consultants of Charleston
Charleston, South Carolina
Rama D. Jager, MD
Attending Physician/Partner
University Retina and Macula Associates
Bedford Park, Illinois
Nancy Kunjukunju, MD
Assistant Professor
School of Medicine
University of Missouri–Kansas City
Assistant Program Director
Department of Ophthalmology
University of Missouri–Kansas City
Kansas City, Missouri
Maria Lozano-Vazquez, MD
Ophthalmology Attending
Department of Surgery
University of Santiago de Compostela
Santiago de Compostela, Spain
Ophthalmology Attending
Department of Surgery
Hospital Naval
Ferral, Spain
Mariana Mata-Plathy, MD
Glaucoma Specialist
Glaucoma Service
Clinica Oftalmologica El Viñedo
Valencia, Carabobo State, Venezuela
Amelia Nelson
Student
Retina Consultants of Charleston
Charleston, South Carolina
Maximiliano Olivera, MD
Assistant Lecturer
Molecular and Cell Medicine
Facultad de Ciencias Biomédicas
Universidad Austral
Pilar, Buenos Aires, Argentina
Veronica Oria, MD
Retina and Vitreous Service
Clínica Oftalmológica El Vinedo
Valencia, Carabobo State, Venezuela
Carlos E. Ortiz, BS
Retina Consultants of Charleston
Charleston, South Carolina
Gabriela Papa-Oliva, MD
Associate Professor
Department of Ophthalmology
Hospital Miguel Pérez Carreño. IVSS
Caracas, Venezuela
Samir Patel, MD
Co-founder and President
Ophthotech Corporation
New York, New York
Hugo Quiroz-Mercado, MD
Director of Ophthalmology
Vitreo-Retina Specialist
Denver Health Medical Center
Professor of Ophthalmology
School of Medicine
University of Colorado
Denver, Colorado
Scott D. Schoenberger, MD
Instructor
Department of Ophthalmology
Vanderbilt University Medical Center
Nashville, Tennessee
Kenneth A. Sharpe, MD
Partner
Retina Consultants of Charleston
Charleston, South Carolina
Veeral Sheth, MD
Clinical Assistant Professor
Department of Ophthalmology
University of Illinois at Chicago
Chicago, Illinois
Director
Scientific Affairs
University Retina and Macula Associates
Lemont, Illinois
Jason S. Slakter, MD
Clinical Professor
Department of Ophthalmology
School of Medicine
New York University
Partner
Vitreous–Retina–Macula Consultants of New York
New York, New York
Robin A. Vora, MD
Fellow, Medical Retina
Department of Ophthalmology
New England Eye Center
Boston, Massachusetts
Attending Physician
Department of Ophthalmology
Kaiser Permanente
Oakland, California
Andre J. Witkin, MD
Retina Fellow
Retina Department
Wills Eye Institute
Philadelphia, Pennsylvania
Lawrence Yannuzzi, MD
Professor of Clinical Ophthalmology
Columbia University Medical School
Founder and Chairman
Vitreous–Retina–Macula Consultants of New York
New York, New York
FOREWORD
We should not forget our mentors, those who went before us and influenced us to
our own personal successes. Just as we are busy and our schedules are saturated
with patients, surgeries, meetings, and the like, so, too, our teachers lived and
flourished. Somehow, they managed to find time to guide and teach us. Those
who influenced me were and are personally involved with the current status of
the diagnosis and treatment of age-related macular degeneration.
In 1984, Drs. Stanley Chang, Harvey Lincoff, and D. Jackson Coleman
walked the eighth floor of Cornell University Medical School and the New York
Hospital and took me under their wing. These iconic figures in the world of
vitreoretinal diseases taught me and countless other students, residents, and
fellows the great balance of clinical practice and significant research.
Stephen J. Ryan served as chairman of the Los Angeles County Hospital
department of ophthalmology and president of the Doheny Eye Institute. During
my residency, he had put together a world class group of retina and macula
specialists, including Peter E. Liggett, John Lean, Edgar Thomas, and others.
They all served as investigators in the Macular Photocoagulation Studies that
were to provide important clinical guidelines for years to follow. Coresidents
Baruch Kuppermann, Pravin Dugel, Victor Gonzales, Keith Pince, Vinh Tran,
Colin Ma, David Wagner, James Tsai, and others immersed themselves in this
great clinical pearl, called the L.A. County Hospital.
Marvin Sears and Peter Liggett provided the leadership and mentorship
during my fellowship at the Yale Eye Center. Marvin was proudest of his great
teaching skills and his publication in Science that showed for the first time the
presence of prostaglandins in the eye.
Age-Related Macular Degeneration represents 2 years of hard work by my
fellow editors and authors. Our plan was to author, edit, and publish a body of
work that would be a source of salient information for our students and
colleagues. Each book chapter has been written to stand alone as a significant
and inclusive text that the reader can use to learn and understand the topic at
hand. It is our hope that patients around the world will benefit from our labor, as
their doctors read and study this book.
D. Virgil Alfaro III, MD
Charleston, South Carolina
ACKNOWLEDGMENTS
The editors are indebted to the hard work and dedication of Sarah M. Granlund,
who served as our project manager during all phases of this textbook. Indeed,
her great diplomacy and administrative skills provided the editorial team with
someone to turn to for help to bring this project to completion.
CONTENTS
Dedication
Contributors
Foreword
Preface
Acknowledgments
17 Lucentis (Ranibizumab)
JoAnna L. Goulah, D. Virgil Alfaro III, Carlos E. Ortiz
27 Retinal Transplantation
Antonio López Bolaños
Index
SECTION 1
INTRODUCTION
Francisco Buzzi in Milan, Italy, was the first to anatomically define the macula
at the end of the 18th century (1782–1784). He described it as the yellow portion
of the posterior retina, lateral to the optic nerve, with a depression in its center.
Also in the 18th century (1797), Fragonard, a French ophthalmologist, made a
very detailed description of the foveal zone, but he failed to mention the central
foveola. In addition, Soemmering (1795–1798) described the macula lutea;
however, he thought that the foveola was a hole or foramen (foraminulum
centrale retinae) and correlated it to the blind spot of the visual field. Finally,
Michaelis in 1838 established the role of the macula, and it was confirmed by
Müller in 1856. At the end of the 19th century, Tratuferi in Italy showed the first
schematic drawings with the topographic localization of the retinal layers. The
relationship between the retinal axonal layer, cones, and rods was established by
Ramon y Cajal in 1894 (1).
The macula is recognized as the specialized region of the retina in charge of
high-resolution visual acuity. Anatomically, it can be defined as the central part
of the posterior retina that contains xanthophyll pigment and two or more layers
of ganglion cells. In this chapter, we focus primarily on the aspects of the normal
anatomy of the macula as an introduction to the understanding and management
of age-related macular degeneration.
EMBRYOLOGY
The neural components of the eye are an extension of the forebrain, and thus part
of the central nervous system (Table 1.1).
aThe marginal layer free from nuclei (not included) appears between the 4th and 5th wk, after which it
disappears. In addition, the transient layer of Chievitz (not included) appears between the 6th wk and 3rd
mo and disappears after the 3rd mo.
The Neuroretina
The embryogenesis of the neuroretina occurs during the first month of life. The
forebrain consists of a single layer of neuroectodermal cells. The optic vesicle
extends laterally from the forebrain and then invaginates to form the optic cup.
There is a double layer of neuroectodermal cells in the optic cup; the apices are
together and basal aspects apart. The macular area appears at the end of the 4th
week.
The inner layer of neuroectoderm becomes the sensory retina posteriorly.
Part of the inner limiting membrane is the basement membrane of the sensory
retina, next to the vitreous. The foveal pit forms late in embryonic life, and
morphologic maturity does not occur until the age when it is possible to obtain
20/20 visual acuity in small children (2). The induction of the foveal pit requires
a normal pigment epithelium (3). The inner layer of neuroectoderm is divided
into the inner neuroblastic layer (Müller, amacrine, and ganglion cells) and the
outer neuroblastic layer (cone and rod, horizontal, and bipolar cells). Between
the inner neuroblastic layer and the outer neuroblastic layer is the transient fiber
layer of Chievitz; this layer will progressively disappear.
The central and peripheral retinae start to differentiate between the first and
third months. The ganglion cell layer becomes thicker, as do the inner plexiform
layer and the amacrine cell layer. The cones appear in the 5th month. They are a
protoplasmic extension of the outer neuroblastic layer (4–6). The rods appear in
the 6th month (5). The macula becomes thinner in the 7th month because the
cells of the different layers move laterally, and the foveal pit appears more
evident (Fig. 1.1) (7,8).
Figure 1.1 Diagram showing the development of the human fovea. A. Human fovea at birth. B. Human
fovea at 45 months. C. Human fovea at 72 years of age. Black lines mark the width of the rod-free foveola;
at birth, the foveola is wider (A) than at 45 months (B). Full foveal development is not complete until
sometime between 15 and 45 months of age.
Vascularization
Vascularization of the retina begins during the 16th week of gestation at the optic
nerve head (12) and normally reaches the ora serrata nasally by term. It is not
quite complete temporally at term because the distance from the optic nerve head
to the ora is greater.
There are differing theories as to how vascularization of the macula occurs.
Some have proposed that the capillary-free zone (CFZ) develops by regression
of previously formed capillaries, because the retina is thin enough in this area
that sufficient oxygen can diffuse inward from the choroidal circulation (12).
Others believe that the CFZ forms primarily, with the embryonic vessels
gradually encircling the center of the fovea and with no evidence of regression
(13).
The Choroid
In the first phase, at 4 weeks, the choroid begins to take form in the
undifferentiated mesenchyma surrounding the optic cup. At 5 to 6 weeks,
endothelial tubes near the pigment epithelium differentiate into capillaries.
Simultaneously, the Bruch's membrane and the vortex veins appear. With the
appearance of the posterior ciliary arteries at 8 weeks, the choriocapillaris is
fully established as a discrete layer.
During the growth phase in the 3rd month, the anterior capillaries arrange
themselves in a linear and radial pattern. The anterior supply of the
choriocapillaris is complete by 3 months, even though the ciliary body and iris
are not yet formed. The capillaries drain into the supra- and infraorbital venous
plexi.
The second phase of choroidal growth includes the formation of large
vessels (Haller's layer) at 4 months. The third phase begins around the 5th month
and is characterized by the formation of medium-sized vessels posterior to the
equator. Anterior to the equator, there are only two layers of vessels, the
choriocapillaris and medium-sized vessels.
Pigmentation of the choroidal melanocytes commences in the 5th month and
continues until after birth, beginning outward near the sclera and progressing
inward toward the Bruch's membrane. The peripapillary, intrascleral vascular
circle of Haller and Zinn develops between 3 and 6 months. Between the 6th and
10th month, the major arterial circle of the iris forms recurrent arterial branches
that supply the anterior choroid during its rapid growth phase (14).
TOPOGRAPHIC ANATOMY
The terms macula and fovea are used in different ways by the anatomist and the
clinician. Anatomically, the macula (macula lutea or central retina) is defined as
the portion of the posterior retina that contains xanthophyll and two or more
layers of ganglion cells. This region is about 5.5 mm in diameter and is centered
approximately 4 mm temporal and 0.8 mm inferior to the center of the optic disk
(15). It corresponds clinically to the posterior pole and is approximately bounded
by the superior and inferior temporal vascular arcades. On the basis of
microscopic anatomy, the macular area can be further subdivided into several
zones (Fig. 1.2).
Figure 1.2 Topographic anatomy of the normal macula. On the basis of microscopic anatomy, the macular
area can be further subdivided into several zones. I. Fovea containing the foveola (1). II. Parafovea. III.
Perifovea.
CLINICAL APPEARANCE
The anatomic subdivisions of the macula are ill-defined ophthalmoscopically.
The margins of either the 0.35-mm diameter foveola or the 1.5-mm diameter
fovea are difficult to define. A poorly defined zone of greater pigmentation (one-
fourth to one disk diameter in size) corresponds to the center of the macula,
which is maximum in the foveolar area. The foveal reflex is present in most
normal eyes, and it lies just in front of the center of the foveola (Fig. 1.3). We
can see the foveal depression with the narrow slit lamp beam.
Figure 1.3 Clinical appearance of the normal macula. A. Fundus in a young patient. B. Fundus in an adult
patient. C. Fundus in a moderately myopic patient. D. Tessellated fundus in an older patient. Large
choroidal vessels are visible in the macular area in (D) because of relative hypopigmentation of the RPE.
(B and D courtesy of Dario Fuenmayor-Rivera and Enrique Murcia.)
The central retinal artery supplies the inner half of the retina. It usually
divides into a superior and inferior trunk within the optic nerve head. These
trunks divide into a nasal branch and a temporal branch. The corresponding
retinal venous branches have much the same distribution as the arteries. They
give off arteriolar and venular branches that posteriorly occur primarily at right
angles to the parent vessel. They divide in a dichotomous way as they course
peripherally. The right-angle branches are referred to as first-order arterioles and
venules. One or more cilioretinal arteries derived from the ciliary circulation
supply the papillomacular area in approximately 20% of patients (17), and
occasionally the entire macula (Fig. 1.4). The blood vessel walls are normally
transparent (16,18).
Figure 1.4 FA shows a cilioretinal artery. (Courtesy of Dario Fuenmayor-Rivera and Enrique Murcia.)
GROSS ANATOMY
The retina loses its normal transparency within hours after death. Xanthophyll is
a yellow pigment made up of two carotenoids (zeaxanthin and lutein) (19,20). It
is apparent in the center of the macula and is highly concentrated in the foveolar
area. The maximal concentration of xanthophyll pigment is in the outer nuclear
and outer plexiform layers. However, we can find xanthophyll within the inner
plexiform layer inside the foveal area (19,21–23). The greatest concentration of
pigment is in the cones' central axons (23).
The entrance site for the short and long posterior ciliary arteries can be
visualized after removal of the choroid. There is no retinal circulation at the
foveola. The short posterior ciliary arteries are concentrated in the macular area
along the temporal margin of the fovea and the peripapillary area. The temporal
long posterior ciliary artery and ciliary nerve enter about one and one-half disk
diameters temporal to the center of the fovea (16).
HISTOLOGY
In the macula, we find the thickest portion of the retina surrounding the thinnest
portion, the foveolar area. At the umbo, the neural retina consists only of the
inner limiting membrane, the Henle's fiber layer, the outer nuclear layer, the
external limiting membrane, and the photoreceptors' outer and inner segments
(Fig. 1.5). The typical neural–retinal histology is established out of the foveola
(Fig. 1.6). The neural retina consists of the inner limiting membrane, the nerve
fiber layer (NFL), the ganglion cell layer, the inner plexiform layer (synaptic
processes between bipolar cells and ganglion cells), the inner nuclear layer
(nuclei of bipolar, horizontal, amacrine, and Müller cells), the outer plexiform
layer (synaptic processes between bipolar cells and photoreceptors), the outer
nuclear layer (nuclei of the rods and cones), the external limiting membrane
(formed by cell junctions between photoreceptors and the terminal optical
processes of Müller cells), and the photoreceptor layer (rod and cone cells). The
bipolar cells, ganglion cells, fibers of the outer plexiform layer (Henle's fiber
layer), and Müller cells are displaced circumferentially and show an oblique
orientation in the macula that causes thickening of the marginal zone and a
central thinning of the retina, forming the fovea.
Figure 1.5 Histology of the normal macula. CFZ, capillary-free zone; f, foveola; u, umbo.
(Microphotograph courtesy of Dario Savino-Zari.)
Figure 1.6 Schematic representation of the human retina illustrating its organization into discrete layers.
AC, amacrine cell; BC, bipolar cell; BM, Bruch's membrane; C, cone photoreceptor; ELM, external
limiting membrane; GC, ganglion cell; HC, horizontal cell; ILM, internal limiting membrane; M, Müller
cell; NFL, nerve fiber layer; R, rod photoreceptor; RPE, retinal pigment epithelium.
Müller cells are modified glial cells. They span the region from the internal
limiting membrane to the external limiting membrane, and they give support to
the neural elements of the retina. The internal limiting membrane consists of a
basement membrane, which is a surface modification of the vitreous body, and
the expanded vitreal processes of Müller cells. This membrane is relatively thick
in the macular region except in the area of the foveola. The internal limiting
membrane serves as an anchoring structure for the collagen framework of the
vitreous. The outer limiting membrane is formed by junctional complexes
between cell membranes of the Müller cells and inner segments of
photoreceptors. Müller cells are connected to the visual cells by a system of
terminal bars (24,25). These junctional complexes probably provide at least a
partial barrier to the passage of large molecules in either direction.
The retinal blood vessels supply the inner half of the retina. The major
branches of the retinal arterial system have the structure of small arteries,
persisting even beyond the equator (15). Retinal arteries do not have an internal
elastic lamina; however, they have a well-developed muscularis (five to seven
layers of smooth muscle cells posteriorly; one or two layers peripherally). Near
the optic disk, the retinal veins have three to four layers of smooth muscle cells,
and after a short distance, the muscle cells are replaced by fibroblasts. There is
controversy concerning the pattern of distribution of the capillary network in the
retina (diffuse arrangement or a two- or three-tier arrangement) (15,26,27). The
superficial network is predominantly postarteriolar and the deep network
prevenular. There is a distinct radial peripapillary capillary network; this
network richly interconnects with the inner retinal capillary layer (27). The large
arterioles and venules of the retinal circulation travel in the NFL and ganglion
cell layer. Close to the CFZ (0.4–0.5 mm in diameter), the capillaries form a
single layer, but elsewhere, the capillaries are present in two or more layers and
extend into the inner nuclear layer. The CFZ is normally vascularized during
prenatal development of the retina. This vascularization undergoes spontaneous
capillary obliteration just before or shortly after birth, forming the CFZ (28).
The foveola is composed entirely of cones; the central 100 μm of the foveola
contains only red and green cones (29). The peak foveal cone density averages
nearly 200,000 per mm2 and falls rapidly with increasing eccentricity, such that
cone density decreases nearly 10-fold within 1 mm of the umbo. Blue cone
density is highest in a zone between 100 and 300 μm from the center of the
fovea. The foveal cones take on a more rodlike shape; blue cones in the macula
tend to have inner segments 10% taller and a more cylindrical shape than their
red and green counterparts (29). Rod cells differ from cones, with their outer
segments consisting of stacks of flattened membrane disks that are separate from
the plasma membrane.
The RPE is a monolayer of hexagonal cells densely adherent to one another
by a system of tight cellular junctions or terminal bars that make up the outer
blood-retinal barrier, which maintains the subretinal space in a state of
deturgescence. In the fovea, there are 30 cones per RPE cell, while in the
periphery, there are 22 rods per RPE cell. Interdigitation of the RPE cells with
the rod and cone outer segments provides only a tenuous adhesion of the RPE to
the sensory retina. The RPE cells in the macular region are taller and contain
increased amounts of melanin pigment than elsewhere (30,31). There is an
inverse relationship between melanin and lipofuscin pigment concentration in
the RPE. Lipofuscin concentration increases initially during the first two decades
of life, and then again in the sixth decade. The concentration of lipofuscin in the
RPE is significantly greater in light- than dark-skinned persons, whereas the
concentration of melanin in the pigment epithelium is similar in light- and dark-
skinned persons. The melanin content of the pigment epithelium and choroidal
melanocytes decline with age (Fig. 1.3).
In young and middle-aged individuals, the RPE is tightly adherent to the
underlying Bruch's membrane by means of its own basement membrane. This
adherence decreases with advancing age. The Bruch's membrane consists of the
basement membrane of the RPE, the inner collagenous layer, an elastic layer, an
outer collagenous layer, and the basement membrane of the choriocapillaris.
Because of its porous structure, it probably plays a minimal role in regulating
movement of substances across it.
The choroid is the posterior aspect of the uveal tract; it is supplied by the
short ciliary arteries, and they are concentrated in the macula and peripapillary
region. These arteries form a rich anastomotic network that quickly empties
large quantities of blood into the choriocapillaris (sinusoidal network). The
choriocapillaris is fenestrated and arranged in a lobular pattern, with a feeding
arteriole in the center of each lobule and several venules peripherally (Fig. 1.7)
(32–40).
Figure 1.7 Schematic representation of the lobular pattern of the choriocapillaris. Each lobule is supplied
by an arteriole. BM, Bruch's membrane; CA, choroidal artery; CV, choroidal vein; RPE, retinal pigment
epithelium.
BLOOD–RETINAL BARRIERS
Retinal perfusion is accomplished by a nonoverlapping, dual system of blood
circulation. For each system, there is a blood–retinal barrier analogous to the
blood–brain barrier. Both barriers confine even relatively small molecules,
because of a nonleaky tight junction between cells (44).
The outer blood–retinal barrier is constituted by the RPE; it blocks the
inward migration of small molecules from the choriocapillaris into the subretinal
space. Anatomically, these junctions include a zonula adherens and adjacent
zonula occludens of the RPE, both situated near the apex of the cell and
encircling it (45). The inner blood–retinal barrier is the retinal vascular
endothelium, including the capillary endothelium. The site of the barrier is the
specialized tight junctions (zonulae occludentes) between individual endothelial
cells (44,46).
NORMAL FLUORESCEIN
ANGIOGRAPHIC FINDINGS
Fluorescein angiography (FA) was developed in the 1950s as a means of
studying vascular flow. FA is still used for this purpose, but it provides much
additional information. Sodium fluorescein is excited by a blue light (465–490
nm), and it emits a fluorescent yellow-green light (peak wavelength of 520–530
nm). Because of its molecular weight (376 kDa), it diffuses freely out of all the
body capillaries except the retina. Approximately 80% of the dye is bound to
plasma proteins (albumin), and it is the unbound fluorescein that is detected
angiographically (16).
FA is performed using a fundus camera. Filters are used to produce the
exciting wavelength. The FA is photographed digitally or with black and white
film because of its superior resolution and speed. On the positive images,
fluorescein appears white and nonfluorescent areas appear black (46). The
normal FA shows the dual nature of the retinal circulation. The larger choroidal
vessels fill first, with almost immediate filling of the choriocapillaris. Rapid
perfusion of the choroid and leakage of the dye from the choriocapillaris give the
fairly uniform background fluorescence.
The tight junctions of the retinal pigment epithelial cells (the outer blood–
retinal barrier) block the fluorescein that leaks from the choriocapillaris and
diffuses through the Bruch's membrane. Normally, the fluorescein does not gain
access to the subsensory retinal space. The retinal vessels, including the
capillaries (the inner blood–retinal barrier), normally do not leak fluorescein.
Thus, the angiogram evaluates both of the blood–retinal barriers (44).
The normal macular region is hypofluorescent or has a barely visible
fluorescence because of the greater density of the RPE in this area and the
presence of xanthophyll in the outer retinal layers. In darkly pigmented
individuals, the increased density of choroidal melanocytes in the macula helps
obscure the background choroidal fluorescence. In addition, the capillaries
appear particularly distinct, because there is only one capillary layer surrounding
the foveal avascular zone (47). Only in the extramacular area, before the
arteriovenous phase, can details of perfusion of the larger choroidal vessels be
detected.
The FA consists of five phases (Fig. 1.8):
Figure 1.8 FA of normal fundus. A. Red-free photograph. B. Arterial phase. C. Early arteriovenous phase.
D. Intermediate arteriovenous phase. E. Venous phase. F. Recirculation phase. (Courtesy of Dario
Fuenmayor-Rivera and Enrique Murcia.)
1. Prearterial phase: The choroid and choriocapillaris fill with dye. A cilioretinal
vessel, if present, usually fills at the same time as the choroidal circulation,
before fluorescein is detectable in the other retinal vessels, approximately 1
second before that of the proximal branches of the central retinal artery (Fig.
1.4).
2. Arterial phase: Lasts until the arteries are completely filled.
3. Arteriovenous phase: Characterized by complete filling of the arteries and
capillaries and the first evidence of laminar flow in the veins.
4. Venous phase: Begins as the arteries are emptying and persists until the veins
are filled with dye.
5. Recirculation phase: Follows the venous phase and represents the first return
of blood to the eye after fluorescein has passed through the kidneys. During
the recirculation phase, the outer edges of the major retinal vessels appear
relatively hyperfluorescent because of the greater amount of fluorescein in the
tangential section of the plasma cuff near the edge of the blood vessels.
Figure 1.9 Indocyanine green video angiography (ICG-V) of the normal fundus. (Courtesy of Dario
Fuenmayor-Rivera.)
NORMAL FUNDUS
AUTOFLUORESCENCE IMAGING
Autofluorescent imaging of the ocular fundus relies on the stimulated emission
of light from molecules, chiefly lipofuscin, in the RPE, in the photoreceptor
outer segments, and in the space between the photoreceptor outer segments and
the RPE (72). Currently, there are two ways to acquire fundus autofluorescence
imaging: one is using a fundus camera-based system (73) and the other is by
confocal scanning laser ophthalmoscopy (74).
ACKNOWLEDGMENTS
Supported in part by the Arevalo-Coutinho Foundation for Research in
Ophthalmology, Caracas, Venezuela.
REFERENCES
1. Amalric P. The macula: 50 years of study: from clinical aspects to genetics. Points de Vue.
1998;39:4–20.
2. Hendrickson AE, Yuodelis C. The morphological development of the human fovea. Ophthalmology.
1984;91:603–612.
3. Fulton AB, Albert DM, Craft JL. Human albinism. Light and electron microscopy study. Arch
Ophthalmol. 1978;96:305–310.
4. Leuenberger P. Stereo-ultrastructure of the retina. Comparative transmission and scanning electron
microscope study. Arch Ophthalmol Rev Gen Ophthalmol. 1971;31:813–822.
5. Mann I. Development of the Human Eye. New York: Grune & Stratton; 1964.
6. Duke-Elder S, Cook C. Normal and abnormal development. In: System of ophthalmology, vol 3, part 1.
St. Louis, MO: Mosby; 1963.
7. Streeten BW. Development of the human retinal pigment epithelium and the posterior segment. Arch
Ophthalmol. 1969;81:383–394.
8. Provis JM, Van Driel D, Billson FA, et al. Development of the human retina: patterns of cell distribution
and redistribution in the ganglion cell layer. J Comp Neurol. 1985;22:429–451.
9. Moore KL. The developing human: clinically oriented embryology. 3rd Ed. Philadelphia, PA: WB
Saunders; 1982:413–416.
10. Weiter JJ, Delori FC, Wing GL, et al. Retinal pigment epithelial lipofuscin and melanin and choroidal
melanin in human eyes. Invest Ophthalmol Vis Sci. 1986;27:145–152.
11. Duvall J. Structure, function, and pathologic responses of pigment epithelium: a review. Semin
Ophthalmol. 1987;2:130–140.
12. Garner A. Retinal angiogenesis: mechanism in health and disease. Semin Ophthalmol. 1987;2:71–80.
13. Engerman RL. Development of the macular circulation. Invest Ophthalmol. 1976;15:835–840.
14. Federman JL, Gouras P, Schubert H, et al. Retina and vitreous. In: Podos SM, Yanoff M, eds. Textbook
of ophthalmology, vol 9. London, UK: Gower Medical; 1994.
15. Hogan MJ, Alvarado JA, Weddell JE. Histology of the human eye: an atlas and textbook. Philadelphia,
PA: WB Saunders; 1971:508–519.
16. Gass JD. Stereoscopic atlas of macular diseases: diagnosis and treatment, 4th Ed, vol 1. St. Louis,
MO: Mosby-Year Book Inc.; 1997:1–17.
17. Justice J Jr, Lehmann RP. Cilioretinal arteries: a study based on review of stereo fundus photographs
and fluorescein angiographic findings. Arch Ophthalmol. 1976;94:1355–1358.
18. Michaelson IC. Retinal circulation in man and animals. Springfield, IL: Charles C. Thomas; 1954.
19. Wald G. Human vision and the spectrum. Science. 1945;101:653–658.
20. Bone RA, Landrum JT, Hime GW, et al. Stereochemistry of the human macular carotenoids. Invest
Ophthalmol Vis Sci. 1993;34:2033–2040.
21. Nussbaum JJ, Pruett RC, Delori FC. Historic perspectives: macular yellow pigment; the first 200 years.
Retina. 1981;1:296–310.
22. Snodderly DM, Auran J, Delori FC. Localization of the macular pigment. Invest Ophthalmol Vis Sci.
1979;18:80.
23. Snodderly DM, Auran J, Delori FC. The macular pigment. II. Spatial distribution in primate retinas.
Invest Ophthalmol Vis Sci. 1984;25:674–685.
24. Fine BS. Limiting membranes of the sensory retina and pigment epithelium: an electron microscopic
study. Arch Ophthalmol. 1961;66:847–860.
25. Blanks JC. Morphology and topography of the retina. In: Ryan SJ. Retina, vol 1, 3rd Ed. St Louis, MO:
Mosby; 2001:32–53.
26. Marquardt R. Ein Beitrag zur Topographie and Anatomie der Netzhautgefasse des menschlichen
Auges. Klin Monatsbl Augenheilkd. 1966;148:50–64.
27. Shimizu K, Ujiie K. Structure of ocular vessels. Tokyo, Japan: Igaku Shoin; 1978.
28. Henkind P, Bellhorn RW, Murphy ME, et al. Development of macular vessels in monkey and cat. Br J
Ophthalmol. 1975;59:703–709.
29. Curcio CA, Allen KA, Sloan KR, et al. Distribution and morphology of human photoreceptors stained
with anti-blue opsin. J Comp Neurol. 1991;312:610–624.
30. Tso MOM, Friedman E. The retinal pigment epithelium I. Comparative histology. Arch Ophthalmol.
1967;78:641–649.
31. Green WR. Retina. In: Spencer WH, Ophthalmic pathology: an atlas and textbook, vol 2, 4th Ed.
Philadelphia, PA: W.B. Saunders Company; 1996:667–681.
32. Amalric PM. Choroidal vessel occlusive syndromes—clinical aspects. Trans Am Acad Ophthalmol
Otolaryngol. 1973;77:OP291–OP299.
33. Hayreh SS. The choriocapillaris. Albrecht von Graefes Arch Klin Exp Ophthalmol. 1974;192:165–179.
34. Hayreh SS. Segmental nature of the choroidal vasculature. Br J Ophthalmol. 1975;59:631–648.
35. Hayreh SS. Submacular choroidal vasculature pattern: experimental fluorescein fundus angiographic
studies. Albrecht von Graefes Arch Klin Exp Ophthalmol. 1974;192:181–196.
36. Krey HF. Segmental vascular patterns of the choriocapillaris. Am J Ophthalmol. 1975;80:198–206.
37. Dollery CT, Henkind P, Kohner EM, et al. Effect of raised intraocular pressure on the retinal and
choroidal circulation. Invest Ophthalmol. 1968;7:191–198.
38. Hayreh SS. Recent advances in fluorescein fundus angiography. Br J Ophthalmol. 1974;58:391–412.
39. Perry HD, Hatfield RV, Tso MOM. Fluorescein pattern of the choriocapillaris in the neonatal rhesus
monkey. Am J Ophthalmol. 1977;84:197–204.
40. Ernest JT, Stern WH, Archer DB. Submacular choroidal circulation. Am J Ophthalmol.
1976;81:574–582.
41. Sarna T. Properties and functions of the ocular melanin: a photobiophysical view. J Photochem
Photobiol B. 1992;12:215–258.
42. Tombran-Tink J, Shivaram SM, Chader GJ, et al. Expression, secretion, and age related down
regulation of pigment epithelium-derived factor, a serpin with neurotrophic activity. J Neurosci.
1995;15:4992–5003.
43. Parver LM, Auker CR, Carpenter DO. The stabilizing effect of the choroidal circulation on the
temperature environment of the macula. Retina. 1982;2:117–120.
44. Cunha-Vaz J. The blood-ocular barriers. Surv Ophthalmol. 1979;23: 279–296.
45. Dubai J. Structure function, and pathologic responses of pigment epithelium: a review. Semin
Ophthalmol. 1987;2:130–140.
46. Kincaid MC. Topographic anatomy, histology and fluorescein angiography. In: Grossniklaus HE,
Kincaid MC. Ophthalmology clinics of North America: macular diseases, vol 6. Philadelphia, PA: WB
Saunders Company; 1993:181–189.
47. Iwasaki M, Inomata H. Relation between superficial capillaries and foveal structures in the human
retina. Invest Ophthalmol Vis Sci. 1986;27: 1698–1705.
48. Rabb MF, Burton TC, Schatz H, et al. Fluorescein angiography of the fundus: a schematic approach to
interpretation. Surv Ophthalmol. 1978;22: 387–403.
49. Flower RW, Hochheimer BF. Clinical infrared absorption angiography of the choroid (letter). Am J
Ophthalmol. 1972;73:458.
50. Piccolino FC, Borgia L, Zinicola E, et al. Pre-injection fluorescence in indocyanine green angiography.
Ophthalmology. 1996;103:1837–1845.
51. Hope-Ross M, Yannuzzi LA, Gragoudas ED, et al. Adverse reactions due to indocyanine green.
Ophthalmology. 1994;101:529–533.
52. Bonte CA, Ceuppens J, Leys AM. Hypotensive shock as a complication of indocyanine green injection.
Retina. 1998;18:476–477.
53. Olsen TW, Lim JI, Capone A Jr, et al. Anaphylactic shock following indocyanine green angiography.
Arch Ophthalmol. 1996;114:97.
54. Hyvärinen L, Flower RW. Indocyanine green fluorescence angiography. Acta Ophthalmol (Copen)
1980;58:528–538.
55. Bischoff PM, Flower RW. Ten years experience with choroidal angiography using indocyanine green
dye: a new routine examination or an epilogue? Doc Ophthalmol. 1985;60:235–291.
56. Slakter JS, Yanuzzi LA, Guyer DR, et al. Indocyanine green angiography. Curr Opin Ophthalmol.
1995;6:25–32.
57. Bartsch DU, Weinreb RN, Zinser G, et al. Confocal scanning infrared laser ophthalmoscopy for
indocyanine green angiography. Am J Ophthalmol. 1995;120:642–651.
58. Klein GJ, Baumgartner RH, Flower RW. An image processing approach to characterizing choroidal
blood flow. Invest Ophthalmol Vis Sci. 1990;31: 629–637.
59. Maberly DAL, Cruess AF. Indocyanine green angiography: an evaluation of image enhancement for
the identification of occult choroidal neovascular membranes. Retina. 1999;19:37–44.
60. Flower RW, Hochheimer BF. Indocyanine green dye fluorescence and infrared absorption choroidal
angiography performed simultaneously with fluorescein angiography. Johns Hopkins Med J.
1976;138:33–42.
61. Bischoff PM, Niederberger HJ, Torok B, et al. Simultaneous indocyanine green and fluorescein
angiography. Retina. 1995;15:91–99.
62. Scheider A, Schroedel C. High resolution indocyanine green angiography with a scanning laser
ophthalmoscope (letter). Am J Ophthalmol. 1989;108:458–459.
63. Holz FG, Bellmann C, Rohrschneider K, et al. Simultaneous confocal scanning laser fluorescein and
indocyanine green angiography. Am J Ophthalmol. 1998;125:227–236.
64. Spaide RF, Orlock DA, Herrmann-Delemazure B, et al. Wide angle indocyanine green angiography.
Retina. 1998;18:44–49.
65. Scheider A, Voeth A, Kaboth A, et al. Fluorescence characteristics of indocyanine green in the normal
choroid and in subretinal neovascular membranes. Ger J Ophthalmol. 1992;1:7–11.
66. Scheider A, Neuhauser L. Fluorescence characteristics of drusen during indocyanine green angiography
and their possible correlation with choroidal perfusion. Ger J Ophthalmol. 1992;1:328–334.
67. Yannuzzi LA, Flower RW, Slakter JS, eds. Indocyanine green angiography. St. Louis, MO: Mosby;
1997.
68. Neubauer AS, Priglinger S, Ullrich S, et al. Comparison of foveal thickness measured with the retinal
thickness analyzer and optical coherence tomography. Retina. 2001;21:596–601.
69. Jo YJ, Heo DW, Shin YI, et al. Diurnal variation of retina thickness measured with time domain and
spectral domain optical coherence tomography in healthy subjects. Invest Ophthalmol Vis Sci.
2011;52(9):497–500.
70. Wakitani Y, Sasoh M, Sugimoto M, et al. Macular thickness measurements in healthy subjects with
different axial lengths using optical coherence tomography. Retina. 2003;23:177–182.
71. Puliafito CA, Hee MR, Schuman JS, et al. Optical coherence tomography of ocular diseases.
Thorofare, NJ: SLACK Inc.; 1996.
72. Delori FC, Dorey K, Staurenghi G, et al. In vivo autofluorescence of the ocular fundus exhibits retinal
pigment epithelial lipofuscin characteristics. Invest Ophthalmol Vis Sci. 1995;36:718–729.
73. Schmitz-Valckenberg S, Holz FG, Bird AC, et al. Fundus autofluorescence imaging: review and
perspectives. Retina. 2008;28:385–409.
74. Holz FG. Autofluorescence imaging of the macula. Ophthalmologe. 2001; 98:10–18.
2
ANIMAL MODELS FOR AGE-
RELATED MACULAR
DEGENERATION
MATÍAS E. CZERWONKO • MARIANA INGOLOTTI •
AMELIA NELSON JAMEL L. F. BROWN • D. VIRGIL
ALFARO III
INTRODUCTION
Age-related macular degeneration (AMD) is a late-onset, multifactorial,
neurodegenerative disease of the retina and the leading cause of irreversible
vision loss in the elderly in the Western world (1). It affects 30 to 50 million
individuals, and clinical hallmarks of AMD are observed in at least one-third of
persons over the age of 75 in industrialized countries (2). Animal models, in
particular, have contributed greatly to current knowledge regarding the etiology,
molecular mechanisms, and histologic features of this extremely prevalent
condition.
As we consider animal models of AMD, with the potential for preclinical
investigation, various pathologic and nonpathologic histologic elements must be
considered. In the particular case of AMD, we can roughly divide its typical
features into two types: histologic and functional alterations.
Among the histologic features that have been reported from the eyes of
patients with AMD are the following:
Most murine models are genetically engineered to manipulate genes that are
suspected to cause macular degeneration-like disorders in humans or might be
somehow associated to AMD pathogenesis. Inflammatory genes (Cfh, Ccl2,
Ccr2, and Cx3cr1), oxidative stress–associated genes (Sod1 and Sod2), and
metabolic pathway genes (mcd, Cp, Heph, ApoE, and ApoB100) have been
manipulated to create models with features that are typical of dry AMD. The
immunologically manipulated mouse model is immunized against
carboxyethylpyrrole (CEP), an endogenous biomarker of oxidative stress.
Finally, the naturally occurring mouse models include retinal degeneration
models with mice exposed to nocive and AMD-related conditions and the
senescence-accelerated mouse (SAM) models with systemic and retinal age-
related pathology, including BM thickening, basal deposits, and CNV.
In the following paragraphs, we explain the most important models of each
of the groups mentioned above. Overall, these models contrast with the murine
models of wet AMD that are mostly induced mechanically and are explained
later in this chapter.
Apolipoproteins
Cholesterol and its transporter, apolipoprotein E (ApoE), are major constituents
of sub-RPE deposits in AMD eyes. Based on these findings, the following
murine models were designed and studied:
Interestingly, ApoE4 mice were the most severely affected, to the point of
having sometimes developed CNV. Transgenic ApoE4 mice fed HF-C diets have
also shown to accumulate amyloid b (Ab), a well-known constituent of human
drusen (39). What is more, systemic anti-Aβ immunotherapy was shown to
protect against loss of visual function and retinal damage, suggesting a role for
Ab in the AMD pathogenesis and functional manifestations (40). Ironically,
while the ApoEe4 allele is correlated with relative protection for AMD in
humans, in the mice, the opposite appears to be true (23,41–43). The reason for
this discrepancy between species still remains unclear.
ApoE3-Leiden transgenic mice: ApoE3-Leiden is a dysfunctional form of
ApoE3 that is associated with a dominantly inherited form of familial
dysbetalipoproteinemia and early onset of atherosclerosis (44). With the
intention of investigating the ApoE3-Leiden mouse as an animal model for
retinal extracellular deposits, transgenic mice carrying the ApoE3-Leiden
gene were created. Eyes were obtained from ApoE3-Leiden transgenic mice
on a HF-C diet or on a normal mouse chow diet for 9 months. As controls,
eyes were collected from ApoE knockout mice on the same dietary
regimens. All eyes of the ApoE3-Leiden mice on a HF-C diet exhibited
basal laminar deposit, whereas ApoE3-Leiden mice on normal chow
displayed basal laminar deposits only occasionally. In both cases, however,
the ultrastructural features of these deposits were comparable with those
seen in human eyes, and they all showed immunoreaction with antihuman
ApoE antibodies. Importantly, no deposits were found in any of the control
mice. These results indicate that ApoE3-Leiden mice can be used as an
animal model for the pathogenesis of basal laminar deposits and that a HF-
C diet enhances the accumulation process. Furthermore, this study supports
the previously suggested involvement of dysfunctional ApoE in the
accumulation of extracellular deposits in the human disease (45,46).
ApoB100 transgenic mice: ApoB100 is the major apolipoprotein in LDL
cholesterol. Lipoprotein particles in the BM have been shown to contain
ApoB100. To study how ApoB100 might contribute to the formation of
lipoproteinaceous deposits in AMD, several groups have examined mice
that express human ApoB100. Young ApoB100 mice developed basal
laminar deposits when fed with a HF diet and exposed to oxidative blue-
green light (47). At older ages, ApoB100 mice exposed to normal diets
exhibited BM thickening, loss of the basal infoldings of the RPE, and basal
laminar deposits (48,49). When these animals were additionally fed HF
diets, more basal linear deposits were observed.
Lipoprotein Receptors
LDL receptor (LDLR) knockout mice are incapable of importing cholesterol,
leading to increased plasma cholesterol levels and development of
atheromatosis. Since atherosclerosis is a well-established risk factor for
developing AMD, a murine knockout model with LDLR deficiency was used to
evaluate changes in the BM. As it was expected, mice lacking LDLR exhibited a
degeneration of the BM with accumulation of lipid particles, which was further
increased after fat intake due to elevated blood lipid levels.
Alternatively, alterations in the gene that codes for VLDL receptor (VLDLR)
have also shown some impact on the onset and progression of retinopathies.
Mice with both alleles mutated do not show signs of dyslipidemia but still
develop retinal neovascularization as early as 2 weeks of age (50–52). These
vessels grow toward the subretinal space, and can form choroidal anastomosis or
cause retinal hemorrhages between 30 and 60 days. Deeper analysis of this
model revealed the development of CNV, elevated levels of vascular endothelial
growth factor (VEGF), and dysregulation of the Wnt pathway all of which
suggested VLDLR as a negative regulator of CNV (53). Therefore, modulation
of the VLDLR may have a great potential from a therapeutic perspective.
More recently, polymorphisms in CD36 have also been shown to be
protective factors in AMD (54). CD36 is a scavenger receptor that binds
oxidized LDL (OxLDL) and is mostly expressed in the apical and basolateral
membrane of the RPE (55–57). Mice deficient for CD36 accumulate subretinal
OxLDL even when fed a regular diet (58). Even though lipids in the RPE come
mostly from photoreceptor outer segments and are exocytosed through the base
of the RPE to be eliminated via the choroid, RPE cells can also take up oxidized
lipids from their cell base, suggesting they may be involved in clearing the
subretinal space from such deposits. Laminar deposits in the BM have been
found to contain OxLDL among other compounds. Consistently, CD36
deficiency in mice resulted in both age-associated accumulation of oxidized
lipids and BM thickening. Coherently, treatment of HF-C-fed ApoE null mice
with a CD36 agonist was shown to diminish thickening of the BM notably as
well as to partially lessen photoreceptor atrophy and preserve function (58).
Cathepsin D
Cathepsin D is an aspartic protease that is highly expressed in the RPE and plays
a decisive role in processing photoreceptor outer segments (59,60). Cathepsin D
is secreted as a proenzyme, which accumulation has been linked to RPE
dysfunction (61). To study the mechanism, transgenic mice with a mutant form
of cathepsin have been developed (62). Mice homozygous for the transgene
demonstrated areas of RPE atrophy as early as 9 months of age (63). Between 10
and 18 months old, these mice exhibited areas of RPE hypertrophy,
photoreceptor atrophy, and reduced ERGs. Remarkably, they also displayed
small yellowish spots in the fundus that were similar to basal laminar and basal
linear spots.
OXIDATIVE DAMAGE MODELS
Clinical and experimental evidence supports that chronic oxidative stress is a
primary contributing factor to numerous retinal degenerative diseases, specially
AMD (64–66). Oxidative damage is normally minimized by the presence of a
wide range of antioxidant and efficient repair systems. Unfortunately, aging
intensifies oxidative damage while decreasing antioxidant capacity, thereby
perturbing the oxidative homeostasis. Postmortem eye examinations from AMD
patients have shown signs in line with extensive free radical damage (67).
Consistently, antioxidant-enriched diets have been shown to reduce the
progression from dry to wet AMD (68).
Several mouse models of chronic oxidative stress displayed many of the
most characteristic hallmarks of AMD. These models can be classified into two
groups: those that lack intrinsic antioxidant mechanisms and those where
additional oxidative stress is applied. However, whether oxidative stress is an
etiologic component or if it is just involved in disease progression is still
controversial.
Carboxyethylpyrrole-Adducted Products
Recent evidence has implicated AMD as an immunologically mediated disease
(69). The protein adduct CEP is present in AMD eye tissue and in the blood of
AMD patients at higher levels than found in age-matched non-AMD tissues.
Autoantibodies to CEP are also higher in AMD blood samples than in controls.
It has been suggested that a signal from the outer retina initiates an immune
response in AMD. CEP acts as a reliable biomarker of oxidative stress and is
currently suspected to be one of those signals (70,71).
To test such hypothesis, Hollyfield et al. (72,73) created a mouse model by
immunizing animals with CEP-adducted mouse serum albumin. Essentially, two
groups were compared: a short-term group that received a strong immunologic
challenge over 3 months and a long-term group that was inoculated with a
weaker challenge but over a year. Both groups successfully developed antibodies
to CEP-adducted albumin. At 3 months, the short-term group showed deposition
of complement component C3d in BM, RPE swelling and lysis, sub-RPE
deposits, pyknosis of overlying photoreceptors, and invasion of macrophages.
Long-term counterparts, on the other hand, exhibited a three- to fivefold
thickening of BM. There existed a solid correlation between the levels of CEP-
specific antibodies and the severity of pathologic alterations. Importantly,
although CEP is also proangiogenic via a VEGF-independent pathway (74),
neither the short-term nor long-term group exhibited signs of CNV, confirming
that the utility of this model is limited for dry AMD only.
SOD Models
Superoxide dismutases (SODs) are one of the major antioxidant defense
systems, which consist of three isoforms in mammals: the cytoplasmic SOD
(SOD1), the mitochondrial SOD (SOD2), and the extracellular SOD (SOD3).
All of these isoforms require catalytic metal (Cu or Mn) for their activation (75).
To investigate the role of oxidative stress in AMD pathogenesis, transgenic mice
targeting the SOD oxidative stress recovery pathways have been created and
examined. With some significant variances, knockout mice for different SOD
genes showed typical features of AMD.
SOD1 levels are particularly high in the retina, and its protective role against
oxidative damage is unquestionable. Imamura et al. showed that SOD1 knockout
mice display age-related pathologic changes in the retina reminiscent of AMD as
early as 7 months of age. Among the most frequent findings were the presence
of drusen, the thickening of BM, and the development of CNV (being thereby
suitable for simulating wet forms of AMD as well) (76). However, prior to the 7-
month period, retinas from these animals were absolutely indistinguishable from
age-matched wild-type animals (76). As with many other models, lesions
increased in number and severity with age: At 10 months of age, 86% of mice
had drusen, compared to very few drusen in age-matched wild-type mice (76),
and at a year of age, RPE vacuolization and degenerative changes were noted.
Remarkably, yellowish drusen-like deposits between the RPE and BM stained
for some of the most typical drusen biomarkers such as vitronectin,
carboxymethyl lysine, and tissue inhibitor metalloproteinase 3 (TIMP3) (76,77).
Senescent SOD1 knockout mice demonstrated evidence of necrotic death of cells
in the inner nuclear layer (INL) and reduced ERGs (78).
SOD2 knockout mice have been used as models for dry AMD as well, but
not without some difficulties. Contrary to SOD1, the antioxidant enzyme
manganese superoxide dismutase (MnSOD), encoded by SOD2, is in the
mitochondrial matrix, and polymorphisms in its encoded gene have been
strongly associated with the development of AMD (79). Unfortunately, most
SOD2 knockout mice died of dilated cardiomyopathy by the early age of 10
days, limiting their use as a model for maculopathies (80). Nevertheless, retinal
histology from some of the few that survived over 3 weeks demonstrated
significant central thinning of all retinal layers (81). Justilien et al. (82) managed
to overcome the limitations of this model by transfecting mouse retina and RPE
with an adenovirus that expresses a ribozyme that degrades SOD2, thereby
reducing its levels only locally and diminishing major systemic consequences.
Thickening of BM, degeneration of the RPE, increased autofluorescence,
elevation of A2E levels, and atrophy of the photoreceptors were among the most
remarkable histologic findings.
OXY Rat
The term OXY rats refer to an inbred strain of Wistar rats that were chosen for
susceptibility or resistance to the early development of cataracts when fed with
diets rich in galactose (101). The susceptible line (OXYS) displays high levels of
hydroxyl radical formation and lipid peroxidation causing mitochondrial
oxidative damage and a senescence-accelerated model. As a result, these rodents
show premature aging and suffer age-related conditions such as cataracts,
emphysema, scoliosis, tumors, and myocardiopathy relatively early in life
(101–104). As for the retina, funduscopic analysis on these rats revealed
choriocapillaris and atrophic areas in the RPE at the age of 45 days (105).
Eventually, rats also exhibited thickening of BM, drusen and RPE detachments,
and, by the age of a year, photoreceptor atrophy, decreased ERGs, destruction of
the choriocapillaris with signs of fibrosis, and even hemorrhagic detachment of
the retina secondary to neovascularization (101,105,106).
COMPLEMENT CASCADE
CFH Models
CFH negatively regulates the complement system by inhibiting the alternative
pathway either by promoting factor I–mediated inactivation of C3b or by
displacing factor Bb from the C3bBb complex and blocking the formation of C3
convertase (125,126). CFH dysfunction may lead to excessive inflammation and
tissue damage, which may contribute to the pathogenesis of AMD in the retina
(127). Patients and mice lacking functional CFH develop macular drusen similar
to those seen in AMD (128–131). At the age of 2 years, these genetically
engineered mice suffered decreased visual acuity, reduction in rod-driven ERG
a- and b-wave responses, increased subretinal autofluorescence, complement
deposition in the retina, and disorganization of photoreceptor outer segments.
However, though there is an association between CFH and AMD, the CFH
knockout mice do not display many of the hallmark AMD features. Contrary to
other models of AMD, almost 30% of these animals showed a substantial
thinning of their BM. Plus, since CFH itself is a main component of drusen, the
loss of this protein may reduce the volume of sub-RPE deposits.
Apolipoprotein E
Amyloid P component
Various-sized complement component C5
The terminal C5b-9 complement complex
Vitronectin, membrane cofactor protein
Annexins
Crystallins
Immunoglobulins
Young age of onset and inheritance pattern in this model are reminiscent of a
number of early-onset maculopathies in humans, rather than AMD. Umeda et al.
assessed the contribution of early-onset human maculopathy susceptibility genes
in this cynomolgus colony. They found no association between the monkey
phenotype and 13 genes that are known to underlie early macular degeneration
syndromes in humans (162). The dominant inheritance and early onset of this
disease could facilitate production of animals for preclinical therapy
development.
Adult-Onset Macular Degeneration in Rhesus
Monkeys (Macaca mulatta)
Even though natural developments of both dry and wet forms of advanced AMD
are only occasionally found in monkeys, many groups have been capable of
documenting the signs of early to intermediate AMD in older rhesus macaques
(Macaca mulatta) (164,166–175). Funduscopic examination of a closed colony
of rhesus macaques found in Cayo Santiago revealed macular drusen in
approximately half of animals older than 9 years and all specimens older than 25
years (168,176–178). Both histologic and ultrastructural examination set the
presence of small and large drusen within the BM as well as beneath the RPE. In
some cases, RPE cells overlying drusen were atrophic. Among the composites
found in the drusen of cynomolgus macaques, we can remark the following:
The apolipoprotein E
Amyloid P component
Complement component C5
The terminal C5b-9 complement complex
Vitronectin, membrane cofactor protein
Annexins
Crystallins
Immunoglobulins
For further studying, a breeding colony derived from these macaques was
established at the University of Florida in 1994, and equivalent features were
displayed (179). In spite of the much higher occurrence of macular drusen in
young adult to middle-age macaques compared to that observed in humans, the
disease seems to progress more slowly and only occasionally lead to geographic
atrophy or CNV (167,176,178). Contrary to early-onset macular degeneration in
cynomolgus macaques, rhesus macaques appear to have a nonmendelian
inheritance pattern. Francis et al. (169) genotyped a population of rhesus
macaques and found that a susceptibility locus in the rhesus homologue of
human 10q26 was associated with macular phenotype. These genes contain
sequence variants significantly associated with affected status in humans and
macaques. As expected, the LOC387715/ARMS2 gene is present in simians
only, thus corresponding with development of the macula. In contrast to human
studies, this study indicated that only the promoter polymorphism in HTRA1
appears to be significantly associated with the disease phenotype.
ANIMAL MODELS OF WET MACULAR
DEGENERATION
Only about 10% of patients suffering from AMD have the wet type. This form,
known also as exudative or neovascular, is characterized by the formation of a
pathologic CNV responsible for most cases of severe blindness (180).
Bleeding, leaking, and scarring from these abnormal blood vessels
eventually cause irreversible damage to the photoreceptors and rapid vision loss
if left untreated. Yet, studies based on animal models have opened the door
toward a new era as far as treatment, prognosis, and morbidity in wet AMD are
concerned. In fact, it was the better understanding of the molecular basis for this
condition that allowed the replacement of aggressive ablative therapies with
more sophisticated and seductive alternatives (181). Agents that delay
neovascular growth and moderate subretinal hemorrhages are one of the most
hopeful breakthroughs in the field of therapeutics for advance AMD and are
currently under evaluation.
The majority of present models for wet AMD rely on either laser or direct
mechanical injury to the RPE/BM complex or alteration of the RPE and
surrounding environment by external interventions (such as exogenous
compounds injected in the subretinal space). Nonetheless, genetically altered
mice have been gaining popularity among experts, who use these “internal”
interventions to explore angiogenic homeostasis within the RPE–choroidal
interface, specially the equilibrium between proangiogenic VEGF and
antiangiogenic PEDF. The interface of these two approaches has brought several
antiangiogenic agents into clinical use. Even though these studies currently focus
on inhibiting VEGF (182), additional use of PEDF via gene therapy constitutes a
realistic idea and may occur in the future (183).
Even though these exudative AMD models recapitulate many of the clinical
and histologic manifestations of CNV in humans, the time to develop, course of
progression, size, and appearance of the lesion vary significantly among them
(184). These models and the differences between them are going to be discussed
in detail in the following sections.
Pig
Another intermediate laser-induced CNV model is the pig. Saishin et al. (219)
used a laser to create defects in the BM in the pig eye and establish histologic
evidence of CNV in 100% of lesions. Image analysis of histologic sections was
used to evaluate drug delivery for the treatment of the CNV. Kiilgaard et al.
studied xenon laser versus diode laser versus mechanical disruption of the BM in
a pig model of CNV. The authors found histopathologic evidence of CNV in
54%, 83%, and 100% of the animals, respectively (236). The pig eye is
approximately the same size as the human eye, and the retina vascularization is
similar to the human. Like the rabbit, the pig circumvents the cost and ethical
issues of the primate. The utility of the pig is for drug delivery experiments.
However, the pig lacks the advantages of high-throughput, short-duration
experiments as available in rat and mouse models of laser-induced CNV.
Surgically BM Rupture
Trauma to the BM can cause localized fundus areas of hemorrhage and
infarction with subsequent neovascular connections between the retina and
choroid (237). Pioneer studies undertaken by Kiilgaard et al. (236) compared
eyes in pigs that underwent retinal photocoagulation with a xenon lamp and
retinal laser photocoagulation with those exposed to mechanical ruptures of the
BM following surgical debridement of the RPE without damage to the
neuroretina. The results were conclusive: Not only did the surgical method show
higher success rates of CNV induction (100% vs. 83% and 54%), but it has also
produced CNV membranes that were morphologically more similar to those
present in human exudative AMD. Contrary to CNV obtained by using laser or
xenon burns, morphology of surgically induced lesions was not dominated by
retinal gliosis and retinal neovascularization, probably due to a preservation of
the neuroretina.
Also, Lassota et al. (238) found that the original surgical technique could be
optimized by avoiding RPE removal prior to the BM perforation. Compared to
those obtained with the original surgical intervention, CNV membranes induced
by using this method were significantly thicker and had a higher cellular content,
were more richly vascularized, and also exhibited the highest propensity to leak
in fluorescence angiograms. It is important to note that even when the surgical
model offers benefits of reduced neuroretinal damage, the technique is limited
by higher cost and the necessity of a three-port vitrectomy.
Macrophages
Macrophages are believed to have a pivotal role in the development of CNV
(249,250). Subretinal injection of macrophages on wild-type or Ccl2 knockout
mice (251) caused CNV with concomitant fibrosis. Nevertheless, since the
protocol included rupture of the BM with a laser at the site of injection, the
meaning of the findings is indefinite.
GENETICALLY ENGINEERED ANIMAL
MODELS OF CNV
Subretinal VEGF Gene Therapy
VEGF is the quintessential promoter of vasculogenesis and angiogenesis
processes in both CNV models and human wet AMD (252,253). In effect, CNV
membranes removed from AMD patients showed VEGF remarkable
immunoreactivity (254,255). It has been hypothesized, therefore, that
overexpression of VEGF would result in concomitant angiogenesis within the
eye.
Numerous groups have sought to put into practice this theory by giving
subretinal injections of adenovirus vectors expressing VEGF to rodents.
Transgenic expression of VEGF in RPE cells resulted in choroidal vascular
permeability, leukocyte adhesion, and intrachoroidal neovascularization, as
measured by angiography and histology. If the VEGF transgene is expressed in
the photoreceptor layer, new vessels originate from the inner retinal vasculature
within the retina and extend into the subretinal space, where they form clusters
of vessels surrounded by RPE cells (256). Adeno-associated virus–mediated
delivery of PEDF to the subretinal or vitreous space results in significant
reduction of neovascularization resulting from laser injury in mice (223). Plus,
pharmacologic blocking of its signaling has shown results as promising as
consistent by successfully inhibiting laser-induced CNV in experimental animal
models (219,257).
The advantage of these models is the ability to study various biologic
components of CNV by comparison with controls and crossbreeding
experiments. Disadvantages relate to the length of time for the CNV to develop,
the relatively small percentages of eyes that develop CNV, and the small size of
the CNV. These models have supported the concept that the best models of CNV
are those that incorporate physical disruption of the BM into the model.
Ccr2-/Ccl2-Deficient Mice
The Ccr2-/Ccl2-deficient mouse model of AMD has received considerable
attention (154). In this model, transgenic mice deficient in either Ccl2 or Ccr2
fail to recruit macrophages to the area of the RPE and BM. This allows for
accumulation of C5a and IgG, both of which induce VEGF production.
Thorough examination of these transgenic mice showed a CNV incidence of
25% (154). These findings have contributed to the understanding of the
pathophysiology of CNV, specifically regarding to macrophage recruitment.
Although the areas of CNV are very small, recent work has shown that CCR3 is
specifically expressed in choroidal neovascular endothelial cells in either of
these knockout mice (258).
VLDLR-Targeted Mutant
Homozygous strains of mice with targeted mutations for VLDLR gene
developed new blood vessels in the area of the outer plexiform layer of the retina
and choroidal anastomoses by 3 months (51).
ApoE Overexpression
Eyes in hypercholesterolemic mice displayed AMD-like changes in the BM/RPE
complex (35). ApoE4 overexpressing transgenic mice fed with HF-C diet
developed CNV in addition to drusen-like and basal laminar deposits as early as
65 weeks of age (36). The CNV was observed in 19% of male and 18% of
female mice and was demonstrated by several methods.
Retinitis Pigmentosa
RP is an inherited disorder that leads to blindness because of the loss of
photoreceptor cells. It is caused by the mutation of multiple genes that encode
for proteins responsible for the maintenance and function of cones and rods. Its
prevalence is around 1 case per 3,000 to 5,000 individuals (268), and the
inheritance can be sporadic, X-linked, and autosomal recessive or dominant,
being the latest accounted for 25% to 30% of all cases of RP (269).
Since neither successful treatment nor cure has been discovered, exploiting
animal experimentation to develop new strategies may be crucial. The greatest
number of known mutations that cause RP occurs in 32 genes, specially in the
rhodopsin (RHO), retinitis pigmentosa 1 (RP1), and retinitis pigmentosa GTPase
regulator (RPGR) genes. However, no single mutation accounts for more than
10% in related patients (270).
RP pathogenesis and therapy have been studied in a wide range of animals
including rodents, dogs, cats, and pigs. Of those, two animal models described in
literature stand out for their utility, and they differ in their Mendelian
inheritance. RPGR mutations are the most common cause of X-linked retinitis
pigmentosa (XLRP). Most of these mutations are present in ORF15, the purine-
rich terminal exon of the predominant splice variant expressed in retina. A
naturally occurring mouse animal model of XLRP has been recently defined.
The retinal degeneration 9 mice carry a 32-base-pair duplication at ORF15 that
leads to a shift in the reading frame causing a nonsense mutation. Therefore, no
protein is translated, and thus, mice exhibit pigment loss and thinning of the
outer nuclear layer (271).
The second model corresponds to an autosomal dominant retinitis
pigmentosa (adRP) miniature pig that carries the most common human mutation
Pro23His (P23H) RHO. Miniature pigs were chosen instead of domestic pigs
because its smaller size makes it easier to manipulate. Mini pigs with the
Pro23His (P23H) RHO mutation successfully developed the clinical phenotype
of human RP: a dramatic reduction in the scotopic b-wave amplitude in the full-
field ERG, reflecting rod photoreceptor dysfunction, and a subsequent, albeit
delayed, reduction in the photopic b-wave amplitude, reflecting cone
photoreceptor dysfunction along with extensive degeneration of the outer
nuclear and reduced thickness of the INLs. Authors supported this model over
rodents on the grounds of convenient eye size, better access to the subretinal
space improving surgical interventions, and a more appropriate biologic model
(since approximately 14% of the photoreceptors in the pig are cones, compared
to only 1% in mice (272)). Cat and dog models of inherited RP are currently
used in research as well (273,274). However, its restricted genetic information,
the presence of major and minor histocompatibility differences using cell-based
therapies, the societal concern over experimentation in them, and the limited
access to retinal progenitor and stem cells in these species constitute major
limitations that frequently impede their use.
Diabetic Retinopathy
DR is a major microvascular complication of diabetes mellitus and the leading
cause of preventable blindness among the working-aged population with a
significant impact on the world's health systems. DR's most accepted
classification includes a nonproliferative (NPDR) and a proliferative form (PDR)
(275). NPDR, previously called background retinopathy, is determined by the
presence of microaneurysms and dot and blot hemorrhages. According to these
microscopic findings, NPDR can be further subdivided into three stages:
REFERENCES
1. Klein R, et al. Ten-year incidence and progression of age-related maculopathy: the Beaver Dam eye
study. Ophthalmology. 2002;109(10):1767–1779.
2. Gehrs KM, et al. Age-related macular degeneration—emerging pathogenetic and therapeutic concepts.
Ann Med. 2006;38(7):450–471.
3. Sparrow JR, et al. A2E, a fluorophore of RPE lipofuscin: can it cause RPE degeneration? Adv Exp Med
Biol. 2003;533:205–211.
4. Green WR, Enger C. Age-related macular degeneration histopathologic studies. The 1992 Lorenz E.
Zimmerman Lecture. Ophthalmology. 1993;100(10):1519–1535.
5. Green WR. Histopathology of age-related macular degeneration. Mol Vis. 1999;5:27.
6. Sarks SH. Ageing and degeneration in the macular region: a clinico-pathological study. Br J
Ophthalmol. 1976;60(5):324–341.
7. Gerth C. The role of the ERG in the diagnosis and treatment of age-related macular degeneration. Doc
Ophthalmol. 2009;118(1):63–68.
8. Gluck JP, Bell J. Ethical issues in the use of animals in biomedical and psychopharmacological research.
Psychopharmacology (Berl). 2003;171(1):6–12.
9. Rands SA. Inclusion of policies on ethical standards in animal experiments in biomedical science
journals. J Am Assoc Lab Anim Sci. 2011;50(6):901–903.
10. Miziara ID, et al. Research ethics in animal models. Braz J Otorhinolaryngol. 2012;78(2):128–131.
11. Pluhar EB. Experimentation on humans and nonhumans. Theor Med Bioeth. 2006;27(4):333–355.
12. Ringach DL. The use of nonhuman animals in biomedical research. Am J Med Sci.
2011;342(4):305–313.
13. Ding X, Patel M, Chan CC. Molecular pathology of age-related macular degeneration. Prog Retin Eye
Res. 2009;28(1):1–18.
14. Curcio CA, et al. Accumulation of cholesterol with age in human Bruch's membrane. Invest
Ophthalmol Vis Sci. 2001;42(1):265–274.
15. Pauleikhoff D, et al. Aging changes in Bruch's membrane. A histochemical and morphologic study.
Ophthalmology. 1990;97(2):171–178.
16. Sheraidah G, et al. Correlation between lipids extracted from Bruch's membrane and age.
Ophthalmology. 1993;100(1):47–51.
17. Sunness JS, et al. Peripheral retinal function in age-related macular degeneration. Arch Ophthalmol.
1985;103(6):811–816.
18. Sunness JS, et al. Retinal sensitivity over drusen and nondrusen areas. A study using fundus perimetry.
Arch Ophthalmol. 1988;106(8):1081–1084.
19. Seddon JM, et al. Progression of age-related macular degeneration: association with body mass index,
waist circumference, and waist-hip ratio. Arch Ophthalmol. 2003;121(6):785–792.
20. Klein R, et al. The association of cardiovascular disease with the long-term incidence of age-related
maculopathy: the Beaver Dam Eye Study. Ophthalmology. 2003;110(6):1273–1280.
21. Tomany SC, et al. Risk factors for incident age-related macular degeneration: pooled findings from 3
continents. Ophthalmology. 2004;111(7):1280–1287.
22. Hyman L, et al. Hypertension, cardiovascular disease, and age-related macular degeneration. Age-
Related Macular Degeneration Risk Factors Study Group. Arch Ophthalmol. 2000;118(3):351–358.
23. Klaver CC, et al. Genetic association of apolipoprotein E with age-related macular degeneration. Am J
Hum Genet. 1998;63(1):200–206.
24. Simonelli F, et al. Apolipoprotein E polymorphisms in age-related macular degeneration in an Italian
population. Ophthalmic Res. 2001;33(6):325–328.
25. Cousins SW, et al. The role of aging, high fat diet and blue light exposure in an experimental mouse
model for basal laminar deposit formation. Exp Eye Res. 2002;75(5):543–553.
26. Chiu CJ, et al. Association between dietary glycemic index and age-related macular degeneration in
nondiabetic participants in the Age-Related Eye Disease Study. Am J Clin Nutr. 2007;86(1):180–188.
27. Chiu CJ, et al. Dietary carbohydrate and the progression of age-related macular degeneration: a
prospective study from the Age-Related Eye Disease Study. Am J Clin Nutr. 2007;86(4):1210–1218.
28. Chiu CJ, et al. Dietary glycemic index and carbohydrate in relation to early age-related macular
degeneration. Am J Clin Nutr. 2006;83(4):880–886.
29. Chiu CJ, Taylor A. Dietary hyperglycemia, glycemic index and metabolic retinal diseases. Prog Retin
Eye Res. 2011;30(1):18–53.
30. Chiu CJ, et al. Informing food choices and health outcomes by use of the dietary glycemic index. Nutr
Rev. 2011;69(4):231–242.
31. Uchiki T, et al. Glycation-altered proteolysis as a pathobiologic mechanism that links dietary glycemic
index, aging, and age-related disease (in nondiabetics). Aging Cell. 2012;11(1):1–13.
32. Weikel KA, et al. Natural history of age-related retinal lesions that precede AMD in mice fed high or
low glycemic index diets. Invest Ophthalmol Vis Sci. 2012;53(2):622–632.
33. Plump AS, et al. Severe hypercholesterolemia and atherosclerosis in apolipoprotein E-deficient mice
created by homologous recombination in ES cells. Cell. 1992;71(2):343–353.
34. Zhang SH, et al. Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein
E. Science. 1992;258(5081):468–471.
35. Dithmar S, et al. Ultrastructural changes in Bruch's membrane of apolipoprotein E-deficient mice.
Invest Ophthalmol Vis Sci. 2000;41(8):2035–2042.
36. Malek G, et al. Apolipoprotein E allele-dependent pathogenesis: a model for age-related retinal
degeneration. Proc Natl Acad Sci U S A. 2005;102(33):11900–11905.
37. Malek G, et al. Initial observations of key features of age-related macular degeneration in APOE
targeted replacement mice. Adv Exp Med Biol. 2006;572:109–117.
38. Sullivan PM, et al. Targeted replacement of the mouse apolipoprotein E gene with the common human
APOE3 allele enhances diet-induced hypercholesterolemia and atherosclerosis. J Biol Chem.
1997;272(29):17972–17980.
39. Mullins RF, et al. Drusen associated with aging and age-related macular degeneration contain proteins
common to extracellular deposits associated with atherosclerosis, elastosis, amyloidosis, and dense deposit
disease. FASEB J. 2000;14(7):835–846.
40. Ding JD, et al. Anti-amyloid therapy protects against retinal pigmented epithelium damage and vision
loss in a model of age-related macular degeneration. Proc Natl Acad Sci U S A. 2011;108(28):E279-E287.
41. Baird PN, et al. The epsilon2 and epsilon4 alleles of the apolipoprotein gene are associated with age-
related macular degeneration. Invest Ophthalmol Vis Sci. 2004;45(5):1311–1315.
42. Schmidt S, et al. A pooled case–control study of the apolipoprotein E (APOE) gene in age-related
maculopathy. Ophthalmic Genet. 2002;23(4):209–223.
43. Souied EH, et al. Macular dystrophy, diabetes, and deafness associated with a large mitochondrial DNA
deletion. Am J Ophthalmol. 1998;125(1):100–103.
44. Havekes L, et al. Apolipoprotein E3-Leiden. A new variant of human apolipoprotein E associated with
familial type III hyperlipoproteinemia. Hum Genet. 1986;73(2):157–163.
45. Kliffen M, et al. The APO(*)E3-Leiden mouse as an animal model for basal laminar deposit. Br J
Ophthalmol. 2000;84(12):1415–1419.
46. van den Maagdenberg AM, et al. Transgenic mice carrying the apolipoprotein E3-Leiden gene exhibit
hyperlipoproteinemia. J Biol Chem. 1993;268(14):10540–10545.
47. Espinosa-Heidmann DG, et al. Basal laminar deposit formation in APO B100 transgenic mice: complex
interactions between dietary fat, blue light, and vitamin E. Invest Ophthalmol Vis Sci. 2004;45(1):260–266.
48. Fujihara M, et al. A human apoB100 transgenic mouse expresses human apoB100 in the RPE and
develops features of early AMD. Exp Eye Res. 2009;88(6):1115–1123.
49. Sallo FB, et al. Bruch's membrane changes in transgenic mice overexpressing the human biglycan and
apolipoprotein b-100 genes. Exp Eye Res. 2009;89(2):178–186.
50. Frykman PK, et al. Normal plasma lipoproteins and fertility in gene-targeted mice homozygous for a
disruption in the gene encoding very low density lipoprotein receptor. Proc Natl Acad Sci U S A.
1995;92(18):8453–8457.
51. Heckenlively JR, et al. Mouse model of subretinal neovascularization with choroidal anastomosis.
Retina. 2003;23(4):518–522.
52. Tiebel O, et al. Mouse very low-density lipoprotein receptor (VLDLR): gene structure, tissue-specific
expression and dietary and developmental regulation. Atherosclerosis. 1999;145(2):239–251.
53. Chen Y, et al. Very low density lipoprotein receptor, a negative regulator of the wnt signaling pathway
and choroidal neovascularization. J Biol Chem. 2007;282(47):34420–34428.
54. Kondo N, et al. Positive association of common variants in CD36 with neovascular age-related macular
degeneration. Aging (Albany NY). 2009;1(2):266–274.
55. Gordiyenko N, et al. RPE cells internalize low-density lipoprotein (LDL) and oxidized LDL (oxLDL)
in large quantities in vitro and in vivo. Invest Ophthalmol Vis Sci. 2004;45(8):2822–2829.
56. Hayes KC, et al. Retinal pigment epithelium possesses both LDL and scavenger receptor activity.
Invest Ophthalmol Vis Sci. 1989;30(2):225–232.
57. Ryeom SW, et al. Binding of anionic phospholipids to retinal pigment epithelium may be mediated by
the scavenger receptor CD36. J Biol Chem. 1996;271(34):20536–20539.
58. Picard E, et al. CD36 plays an important role in the clearance of oxLDL and associated age-dependent
sub-retinal deposits. Aging (Albany NY). 2010;2(12):981–989.
59. Bosch E, Horwitz J, Bok D. Phagocytosis of outer segments by retinal pigment epithelium:
phagosome–lysosome interaction. J Histochem Cytochem. 1993;41(2):253–263.
60. Regan CM, et al. Degradation of rhodopsin by a lysosomal fraction of retinal pigment epithelium:
biochemical aspects of the visual process. XLI. Exp Eye Res. 1980;30(2):183–191.
61. Rakoczy PE, et al. Correlation between autofluorescent debris accumulation and the presence of
partially processed forms of cathepsin D in cultured retinal pigment epithelial cells challenged with rod
outer segments. Exp Eye Res. 1996;63(2):159–167.
62. Zhang D, et al. A model for a blinding eye disease of the aged. Biogerontology. 2002;3(1–2):61–66.
63. Rakoczy PE, et al. Progressive age-related changes similar to age-related macular degeneration in a
transgenic mouse model. Am J Pathol. 2002;161(4):1515–1524.
64. Cruickshanks KJ, et al. The prevalence of age-related maculopathy by geographic region and ethnicity.
The Colorado-Wisconsin Study of Age-Related Maculopathy. Arch Ophthalmol. 1997;115(2):242–250.
65. Vingerling JR, et al. Age-related macular degeneration and smoking. The Rotterdam Study. Arch
Ophthalmol. 1996;114(10):1193–1196.
66. Wenzel A, et al. Molecular mechanisms of light-induced photoreceptor apoptosis and neuroprotection
for retinal degeneration. Prog Retin Eye Res. 2005;24(2):275–306.
67. Nowak M, et al. Changes in blood antioxidants and several lipid peroxidation products in women with
age-related macular degeneration. Eur J Ophthalmol. 2003;13(3):281–286.
68. Age-Related Eye Disease Study Research Group. A randomized, placebo-controlled, clinical trial of
high-dose supplementation with vitamins C and E and beta carotene for age-related cataract and vision loss:
AREDS report no. 9. Arch Ophthalmol. 2001;119(10):1439–1452.
69. Patel M, Chan CC, Immunopathological aspects of age-related macular degeneration. Semin
Immunopathol. 2008;30(2):97–110.
70. Anderson RE, Lipids of ocular tissues. IV. A comparison of the phospholipids from the retina of six
mammalian species. Exp Eye Res. 1970;10(2):339–344.
71. Gu X, et al. Carboxyethylpyrrole protein adducts and autoantibodies, biomarkers for age-related
macular degeneration. J Biol Chem. 2003;278(43):42027–42035.
72. Hollyfield JG, et al. Oxidative damage-induced inflammation initiates age-related macular
degeneration. Nat Med. 2008;14(2):194–198.
73. Hollyfield JG, Perez VL, Salomon RG. A hapten generated from an oxidation fragment of
docosahexaenoic acid is sufficient to initiate age-related macular degeneration. Mol Neurobiol. 2010;41(2–
3):290–298.
74. Ebrahem Q, et al. Carboxyethylpyrrole oxidative protein modifications stimulate neovascularization:
implications for age-related macular degeneration. Proc Natl Acad Sci U S A. 2006;103(36):13480–13484.
75. Behndig A, et al. Superoxide dismutase isoenzymes in the human eye. Invest Ophthalmol Vis Sci.
1998;39(3):471–475.
76. Imamura Y, et al. Drusen, choroidal neovascularization, and retinal pigment epithelium dysfunction in
SOD1-deficient mice: a model of age-related macular degeneration. Proc Natl Acad Sci U S A.
2006;103(30):11282–11287.
77. Crabb JW, et al. Drusen proteome analysis: an approach to the etiology of age-related macular
degeneration. Proc Natl Acad Sci U S A. 2002;99(23):14682–14687.
78. Hashizume K, et al. Retinal dysfunction and progressive retinal cell death in SOD1-deficient mice. Am
J Pathol. 2008;172(5):1325–1331.
79. Kimura K, et al. Genetic association of manganese superoxide dismutase with exudative age-related
macular degeneration. Am J Ophthalmol. 2000;130(6):769–773.
80. Li Y, et al. Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese
superoxide dismutase. Nat Genet. 1995;11(4):376–381.
81. Sandbach JM, et al. Ocular pathology in mitochondrial superoxide dismutase (Sod2)-deficient mice.
Invest Ophthalmol Vis Sci. 2001;42(10):2173–2178.
82. Justilien V, et al. SOD2 knockdown mouse model of early AMD. Invest Ophthalmol Vis Sci.
2007;48(10):4407–4420.
83. Dunaief JL. Iron induced oxidative damage as a potential factor in age-related macular degeneration:
the Cogan Lecture. Invest Ophthalmol Vis Sci. 2006;47(11):4660–4664.
84. Hort GM, et al. Delayed type hypersensitivity-associated disruption of splenic periarteriolar lymphatic
sheaths coincides with temporary loss of IFN-gamma production and impaired eradication of bacteria in
Brucella abortus-infected mice. Microbes Infect. 2003;5(2):95–106.
85. Miyajima H, et al. Familial apoceruloplasmin deficiency associated with blepharospasm and retinal
degeneration. Neurology. 1987;37(5):761–767.
86. Morita H, et al. Hereditary ceruloplasmin deficiency with hemosiderosis: a clinicopathological study of
a Japanese family. Ann Neurol. 1995;37(5):646–656.
87. Yamaguchi K, et al. Retinal degeneration in hereditary ceruloplasmin deficiency. Ophthalmologica.
1998;212(1):11–14.
88. Patel BN, et al. Ceruloplasmin regulates iron levels in the CNS and prevents free radical injury. J
Neurosci. 2002;22(15):6578–6586.
89. Harris ZL, et al. Targeted gene disruption reveals an essential role for ceruloplasmin in cellular iron
efflux. Proc Natl Acad Sci U S A. 1999;96(19):10812–10817.
90. Vulpe CD, et al. Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is
defective in the sla mouse. Nat Genet. 1999;21(2):195–199.
91. Hahn P, et al. Disruption of ceruloplasmin and hephaestin in mice causes retinal iron overload and
retinal degeneration with features of age-related macular degeneration. Proc Natl Acad Sci U S A.
2004;101(38):13850–13855.
92. Hadziahmetovic M, et al. Ceruloplasmin/hephaestin knockout mice model morphologic and molecular
features of AMD. Invest Ophthalmol Vis Sci. 2008;49(6):2728–2736.
93. Evans JR. Risk factors for age-related macular degeneration. Prog Retin Eye Res. 2001;20(2):227–253.
94. Klein R, et al. Further observations on the association between smoking and the long-term incidence
and progression of age-related macular degeneration: the Beaver Dam Eye Study. Arch Ophthalmol.
2008;126(1):115–121.
95. Seddon JM, et al. A prospective study of cigarette smoking and age-related macular degeneration in
women. JAMA. 1996;276(14):1141–1146.
96. Smith W, et al. Risk factors for age-related macular degeneration: pooled findings from three
continents. Ophthalmology. 2001;108(4):697–704.
97. Thornton J, et al. Smoking and age-related macular degeneration: a review of association. Eye (Lond).
2005;19(9):935–944.
98. Smith CJ, Hansch C. The relative toxicity of compounds in mainstream cigarette smoke condensate.
Food Chem Toxicol. 2000;38(7):637–646.
99. Espinosa-Heidmann DG, et al. Cigarette smoke-related oxidants and the development of sub-RPE
deposits in an experimental animal model of dry AMD. Invest Ophthalmol Vis Sci. 2006;47(2):729–737.
100. Pons M, Marin-Castano ME. Cigarette smoke-related hydroquinone dysregulates MCP-1, VEGF and
PEDF expression in retinal pigment epithelium in vitro and in vivo. PLoS One. 2011;6(2):e16722.
101. Salganik RI, et al. Inherited enhancement of hydroxyl radical generation and lipid peroxidation in the
S strain rats results in DNA rearrangements, degenerative diseases, and premature aging. Biochem Biophys
Res Commun. 1994;199(2):726–733.
102. Kolosova NG, et al. OXYS rats as a model of senile cataract. Bull Exp Biol Med.
2003;136(4):415–419.
103. Marsili S, et al. Cataract formation in a strain of rats selected for high oxidative stress. Exp Eye Res.
2004;79(5):595–612.
104. Salganik RI, et al. Impairment of respiratory functions in mitochondria of rats with an inherited
hyperproduction of free radicals. Biochem Biophys Res Commun. 1994;205(1):180–185.
105. Markovets AM, et al. Alterations of retinal pigment epithelium cause AMD-like retinopathy in
senescence-accelerated OXYS rats. Aging (Albany NY). 2011;3(1):44–54.
106. Neroev VV, et al. Mitochondria-targeted plastoquinone derivatives as tools to interrupt execution of
the aging program. 4. Age-related eye disease. SkQ1 returns vision to blind animals. Biochemistry (Mosc).
2008;73(12):1317–1328.
107. Xu H, et al. Age-dependent accumulation of lipofuscin in perivascular and subretinal microglia in
experimental mice. Aging Cell. 2008;7(1):58–68.
108. Xu H, Chen M, Forrester JV. Para-inflammation in the aging retina. Prog Retin Eye Res.
2009;28(5):348–368.
109. van der Schaft TL, et al. Histologic features of the early stages of age-related macular degeneration. A
statistical analysis. Ophthalmology. 1992;99(2):278–286.
110. Spraul CW, Grossniklaus HE. Characteristics of Drusen and Bruch's membrane in postmortem eyes
with age-related macular degeneration. Arch Ophthalmol. 1997;115(2):267–273.
111. Anderson DH, et al. A role for local inflammation in the formation of drusen in the aging eye. Am J
Ophthalmol. 2002;134(3):411–431.
112. Chan CC, et al. Ccl2/Cx3cr1-deficient mice: an animal model for age-related macular degeneration.
Ophthalmic Res. 2008;40(3–4):124–128.
113. Boekhoorn SS, et al. C-reactive protein level and risk of aging macula disorder: the Rotterdam Study.
Arch Ophthalmol. 2007;125(10):1396–1401.
114. Seddon JM, et al. Association between C-reactive protein and age-related macular degeneration.
JAMA. 2004;291(6):704–710.
115. Gold B, et al. Variation in factor B (BF) and complement component 2 (C2) genes is associated with
age-related macular degeneration. Nat Genet. 2006;38(4):458–462.
116. Maller J, et al. Common variation in three genes, including a noncoding variant in CFH, strongly
influences risk of age-related macular degeneration. Nat Genet. 2006;38(9):1055–1059.
117. Montes T, et al. Functional basis of protection against age-related macular degeneration conferred by a
common polymorphism in complement factor B. Proc Natl Acad Sci U S A. 2009;106(11):4366–4371.
118. Reynolds R, et al. Plasma complement components and activation fragments: associations with age-
related macular degeneration genotypes and phenotypes. Invest Ophthalmol Vis Sci.
2009;50(12):5818–5827.
119. Yates JR, et al. Complement C3 variant and the risk of age-related macular degeneration. N Engl J
Med. 2007;357(6):553–561.
120. Edwards AO, et al. Complement factor H polymorphism and age-related macular degeneration.
Science. 2005;308(5720):421–424.
121. Hageman GS, et al. A common haplotype in the complement regulatory gene factor H (HF1/CFH)
predisposes individuals to age-related macular degeneration. Proc Natl Acad Sci U S A.
2005;102(20):7227–7232.
122. Haines JL, et al. Complement factor H variant increases the risk of age-related macular degeneration.
Science. 2005;308(5720):419–421.
123. Thakkinstian A, et al. Systematic review and meta-analysis of the association between complement
factor H Y402H polymorphisms and age-related macular degeneration. Hum Mol Genet.
2006;15(18):2784–2790.
124. Zareparsi S, et al. Strong association of the Y402H variant in complement factor H at 1q32 with
susceptibility to age-related macular degeneration. Am J Hum Genet. 2005;77(1):149–153.
125. Alsenz J, et al. Structural and functional analysis of the complement component factor H with the use
of different enzymes and monoclonal antibodies to factor H. Biochem J. 1985;232(3):841–850.
126. Pickering MC, et al. Uncontrolled C3 activation causes membranoproliferative glomerulonephritis in
mice deficient in complement factor H. Nat Genet. 2002;31(4):424–428.
127. Johnson PT, et al. Individuals homozygous for the age-related macular degeneration risk-conferring
variant of complement factor H have elevated levels of CRP in the choroid. Proc Natl Acad Sci U S A.
2006;103(46):17456–17461.
128. Duvall-Young J, et al. Fundus changes in mesangiocapillary glomerulonephritis type II: clinical and
fluorescein angiographic findings. Br J Ophthalmol. 1989;73(11):900–906.
129. Duvall-Young J, MacDonald MK, McKechnie NM. Fundus changes in (type II) mesangiocapillary
glomerulonephritis simulating drusen: a histopathological report. Br J Ophthalmol. 1989;73(4):297–302.
130. Raines MF, Duvall-Young J, Short CD. Fundus changes in mesangiocapillary glomerulonephritis type
II: vitreous fluorophotometry. Br J Ophthalmol. 1989;73(11):907–910.
131. Leys A, et al. Fundus changes in membranoproliferative glomerulonephritis type II. A fluorescein
angiographic study of 23 patients. Graefes Arch Clin Exp Ophthalmol. 1991;229(5):406–410.
132. Dewan A, et al. HTRA1 promoter polymorphism in wet age-related macular degeneration. Science.
2006;314(5801):989–992.
133. Ufret-Vincenty RL, et al. Transgenic mice expressing variants of complement factor H develop AMD-
like retinal findings. Invest Ophthalmol Vis Sci. 2010;51(11):5878–5887.
134. Cashman SM, et al. Expression of complement component 3 (C3) from an adenovirus leads to
pathology in the murine retina. Invest Ophthalmol Vis Sci. 2011;52(6):3436–3445.
135. Johnson LV, et al. A potential role for immune complex pathogenesis in drusen formation. Exp Eye
Res. 2000;70(4):441–449.
136. Nozaki M, et al. Drusen complement components C3a and C5a promote choroidal neovascularization.
Proc Natl Acad Sci U S A. 2006;103(7):2328–2333.
137. Graves DT, Jiang Y. Chemokines, a family of chemotactic cytokines. Crit Rev Oral Biol Med.
1995;6(2):109–118.
138. Murphy PM, et al. International union of pharmacology. XXII. Nomenclature for chemokine
receptors. Pharmacol Rev. 2000;52(1):145–176.
139. Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity.
2000;12(2):121–127.
140. Gupta N, Brown KE, Milam AH. Activated microglia in human retinitis pigmentosa, late-onset retinal
degeneration, and age-related macular degeneration. Exp Eye Res. 2003;76(4):463–471.
141. van der Schaft TL, et al. Early stages of age-related macular degeneration: an immunofluorescence
and electron microscopy study. Br J Ophthalmol. 1993;77(10):657–661.
142. Raoul W, et al. CCL2/CCR2 and CX3CL1/CX3CR1 chemokine axes and their possible involvement
in age-related macular degeneration. J Neuroinflammation. 2010;7:87.
143. Chan CC, et al. Detection of CX3CR1 single nucleotide polymorphism and expression on archived
eyes with age-related macular degeneration. Histol Histopathol. 2005;20(3):857–863.
144. Combadiere C, et al. CX3CR1-dependent subretinal microglia cell accumulation is associated with
cardinal features of age-related macular degeneration. J Clin Invest. 2007;117(10):2920–2928.
145. Tuo J, et al. Murine ccl2/cx3cr1 deficiency results in retinal lesions mimicking human age-related
macular degeneration. Invest Ophthalmol Vis Sci. 2007;48(8):3827–3836.
146. Yang X, et al. Polymorphisms in CFH, HTRA1 and CX3CR1 confer risk to exudative age-related
macular degeneration in Han Chinese. Br J Ophthalmol. 2010;94(9):1211–1214.
147. Kuziel WA, et al. Severe reduction in leukocyte adhesion and monocyte extravasation in mice
deficient in CC chemokine receptor 2. Proc Natl Acad Sci U S A. 1997;94(22):12053–12058.
148. Lu B, et al. Abnormalities in monocyte recruitment and cytokine expression in monocyte
chemoattractant protein 1-deficient mice. J Exp Med. 1998;187(4):601–608.
149. Higgins GT, et al. Induction of angiogenic cytokine expression in cultured RPE by ingestion of
oxidized photoreceptor outer segments. Invest Ophthalmol Vis Sci. 2003;44(4):1775–1782.
150. Jonas JB, et al. Monocyte chemoattractant protein 1, intercellular adhesion molecule 1, and vascular
cell adhesion molecule 1 in exudative age-related macular degeneration. Arch Ophthalmol.
2010;128(10):1281–1286.
151. Yamada K, et al. Inhibition of laser-induced choroidal neovascularization by atorvastatin by
downregulation of monocyte chemotactic protein-1 synthesis in mice. Invest Ophthalmol Vis Sci.
2007;48(4):1839–1843.
152. Tsutsumi C, et al. The critical role of ocular-infiltrating macrophages in the development of choroidal
neovascularization. J Leukoc Biol. 2003;74(1):25–32.
153. Luhmann UF, et al. The drusenlike phenotype in aging Ccl2-knockout mice is caused by an
accelerated accumulation of swollen autofluorescent subretinal macrophages. Invest Ophthalmol Vis Sci.
2009;50(12):5934–5943.
154. Ambati J, et al. An animal model of age-related macular degeneration in senescent Ccl-2- or Ccr-2-
deficient mice. Nat Med. 2003;9(11):1390–1397.
155. Cardona AE, et al. Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci.
2006;9(7):917–924.
156. Silverman MD, et al. Constitutive and inflammatory mediator-regulated fractalkine expression in
human ocular tissues and cultured cells. Invest Ophthalmol Vis Sci. 2003;44(4):1608–1615.
157. Combadiere C, et al. Decreased atherosclerotic lesion formation in CX3CR1/apolipoprotein E double
knockout mice. Circulation. 2003;107(7):1009–1016.
158. Jung S, et al. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green
fluorescent protein reporter gene insertion. Mol Cell Biol. 2000;20(11):4106–4114.
159. Ross RJ, et al. Immunological protein expression profile in Ccl2/Cx3cr1 deficient mice with lesions
similar to age-related macular degeneration. Exp Eye Res. 2008;86(4):675–683.
160. Mattapallil MJ, et al. The Rd8 mutation of the Crb1 gene is present in vendor lines of C57BL/6 N
mice and embryonic stem cells, and confounds ocular induced mutant phenotypes. Invest Ophthalmol Vis
Sci. 2012;53(6):2921–2927.
161. Jeffrey BG, Neuringer M. Age-related decline in rod phototransduction sensitivity in rhesus monkeys
fed an n-3 fatty acid-deficient diet. Invest Ophthalmol Vis Sci. 2009;50(9):4360–4367.
162. Umeda S, et al. Early-onset macular degeneration with drusen in a cynomolgus monkey (Macaca
fascicularis) pedigree: exclusion of 13 candidate genes and loci. Invest Ophthalmol Vis Sci.
2005;46(2):683–691.
163. Suzuki MT, Terao K, Yoshikawa Y. Familial early onset macular degeneration in cynomolgus
monkeys (Macaca fascicularis). Primates. 2003;44(3):291–294.
164. Nicolas MG, et al. Studies on the mechanism of early onset macular degeneration in cynomolgus
monkeys. II. Suppression of metallothionein synthesis in the retina in oxidative stress. Exp Eye Res.
1996;62(4):399–408.
165. Nicolas MG, et al. Studies on the mechanism of early onset macular degeneration in cynomolgus
(Macaca fascicularis) monkeys. I. Abnormal concentrations of two proteins in the retina. Exp Eye Res.
1996;62(3):211–219.
166. Bellhorn RW, et al. Pigmentary abnormalities of the macula in rhesus monkeys: clinical observations.
Invest Ophthalmol Vis Sci. 1981;21(6):771–781.
167. Dawson WW, et al. Macular disease in related rhesus monkeys. Doc Ophthalmol.
1989;71(3):253–263.
168. El-Mofty AA, et al. Retinal degeneration in rhesus monkeys. Macaca mulatta. Survey of three
seminatural free-breeding colonies. Exp Eye Res. 1980;31(2):147–166.
169. Francis PJ, et al. Rhesus monkeys and humans share common susceptibility genes for age-related
macular disease. Hum Mol Genet. 2008;17(17):2673–2680.
170. Hope GM, et al. A primate model for age related macular drusen. Br J Ophthalmol. 1992;76(1):11–16.
171. Jonas JB, Hayreh SS, Martus P. Influence of arterial hypertension and diet-induced atherosclerosis on
macular drusen. Graefes Arch Clin Exp Ophthalmol. 2003;241(2):125–134.
172. Monaco WA, Wormington CM. The rhesus monkey as an animal model for age-related maculopathy.
Optom Vis Sci. 1990;67(7):532–537.
173. Olin KL, et al. Trace element status and free radical defense in elderly rhesus macaques (Macaca
mulatta) with macular drusen. Proc Soc Exp Biol Med. 1995;208(4):370–377.
174. Stafford TJ, Anness SH, Fine BS. Spontaneous degenerative maculopathy in the monkey.
Ophthalmology. 1984;91(5):513–521.
175. Ulshafer RJ, et al. Macular degeneration in a community of rhesus monkeys. Ultrastructural
observations. Retina. 1987;7(3):198–203.
176. Dawson WW, et al. Adult-onset macular degeneration in the Cayo Santiago macaques. P R Health Sci
J. 1989;8(1):111–115.
177. El-Mofty A, et al. Macular degeneration in rhesus monkey (Macaca mulatta). Exp Eye Res.
1978;27(4):499–502.
178. Engel HM, et al. Degenerative changes in maculas of rhesus monkeys. Ophthalmologica.
1988;196(3):143–150.
179. Dawson WW, et al. Maculas, monkeys, models. AMD and aging. Vision Res. 2008;48(3):360–365.
180. Klein R, et al. The relationship of age-related maculopathy, cataract, and glaucoma to visual acuity.
Invest Ophthalmol Vis Sci. 1995;36(1):182–191.
181. Group CR, et al. Ranibizumab and bevacizumab for neovascular age-related macular degeneration. N
Engl J Med. 2011;364(20):1897–1908.
182. van Wijngaarden P, Qureshi SH, Inhibitors of vascular endothelial growth factor (VEGF) in the
management of neovascular age-related macular degeneration: a review of current practice. Clin Exp
Optom. 2008;91(5):427–437.
183. Campochiaro PA. Gene therapy for ocular neovascularization. Curr Gene Ther. 2007;7(1):25–33.
184. Edelman JL, Castro MR. Quantitative image analysis of laser-induced choroidal neovascularization in
rat. Exp Eye Res. 2000;71(5):523–533.
185. Bora PS, et al. Immunotherapy for choroidal neovascularization in a laser-induced mouse model
simulating exudative (wet) macular degeneration. Proc Natl Acad Sci U S A. 2003;100(5):2679–2684.
186. Economou MA, et al. Inhibition of VEGF secretion and experimental choroidal neovascularization by
picropodophyllin (PPP), an inhibitor of the insulin-like growth factor-1 receptor. Acta Ophthalmol. 2008;86
Thesis 4:42–49.
187. Semkova I, et al. Investigation of laser-induced choroidal neovascularization in the rat. Invest
Ophthalmol Vis Sci. 2003;44(12):5349–5354.
188. Ciulla MM, et al. Vascular network changes in the retina during ageing in normal subjects: a
computerized quantitative analysis. Ital Heart J. 2000;1(5):361–364.
189. Ryan SJ. The development of an experimental model of subretinal neovascularization in disciform
macular degeneration. Trans Am Ophthalmol Soc. 1979;77:707–745.
190. Tobe T, et al. Targeted disruption of the FGF2 gene does not prevent choroidal neovascularization in a
murine model. Am J Pathol. 1998;153(5):1641–1646.
191. Campa C, et al. Effects of an anti-VEGF-A monoclonal antibody on laser-induced choroidal
neovascularization in mice: optimizing methods to quantify vascular changes. Invest Ophthalmol Vis Sci.
2008;49(3):1178–1183.
192. Seo MS, et al. Dramatic inhibition of retinal and choroidal neovascularization by oral administration
of a kinase inhibitor. Am J Pathol. 1999;154(6):1743–1753.
193. Campos M, et al. A novel imaging technique for experimental choroidal neovascularization. Invest
Ophthalmol Vis Sci. 2006;47(12):5163–5170.
194. Ogata N, et al. Expression of basic fibroblast growth factor mRNA in developing choroidal
neovascularization. Curr Eye Res. 1996;15(10):1008–1018.
195. Yamada H, et al. Cell injury unmasks a latent proangiogenic phenotype in mice with increased
expression of FGF2 in the retina. J Cell Physiol. 2000;185(1):135–142.
196. Oshima Y, et al. Increased expression of VEGF in retinal pigmented epithelial cells is not sufficient to
cause choroidal neovascularization. J Cell Physiol. 2004;201(3):393–400.
197. Shen WY, et al. Expression of cell adhesion molecules and vascular endothelial growth factor in
experimental choroidal neovascularisation in the rat. Br J Ophthalmol. 1998;82(9):1063–1071.
198. Wada M, et al. Expression of vascular endothelial growth factor and its receptor (KDR/flk-1) mRNA
in experimental choroidal neovascularization. Curr Eye Res. 1999;18(3):203–213.
199. Yi X, et al. Vascular endothelial growth factor expression in choroidal neovascularization in rats.
Graefes Arch Clin Exp Ophthalmol. 1997;235(5):313–319.
200. Ando A, et al. Nitric oxide is proangiogenic in the retina and choroid. J Cell Physiol.
2002;191(1):116–124.
201. Berglin L, et al. Reduced choroidal neovascular membrane formation in matrix metalloproteinase-2-
deficient mice. Invest Ophthalmol Vis Sci. 2003;44(1):403–408.
202. Lambert V, et al. Matrix metalloproteinase-9 contributes to choroidal neovascularization. Am J
Pathol. 2002;161(4):1247–1253.
203. Zhu J, et al. Focal adhesion kinase signaling pathway participates in the formation of choroidal
neovascularization and regulates the proliferation and migration of choroidal microvascular endothelial
cells by acting through HIF-1 and VEGF expression in RPE cells. Exp Eye Res. 2009;88(5):910–918.
204. Sakurai E, et al. Macrophage depletion inhibits experimental choroidal neovascularization. Invest
Ophthalmol Vis Sci. 2003;44(8):3578–3585.
205. Dobi ET, Puliafito CA, Destro M. A new model of experimental choroidal neovascularization in the
rat. Arch Ophthalmol. 1989;107(2):264–269.
206. Zhang ZX, et al. Hypoxia specific SDF-1 expression by retinal pigment epithelium initiates bone
marrow-derived cells to participate in Choroidal neovascularization in a laser-induced mouse model. Curr
Eye Res. 2011;36(9):838–849.
207. Bora NS, et al. Complement activation via alternative pathway is critical in the development of laser-
induced choroidal neovascularization: role of factor B and factor H. J Immunol. 2006;177(3):1872–1878.
208. Lima e Silva R, et al. Suppression and regression of choroidal neovascularization by polyamine
analogues. Invest Ophthalmol Vis Sci. 2005;46(9):3323–3330.
209. Ciulla TA, et al. Intravitreal triamcinolone acetonide inhibits choroidal neovascularization in a laser-
treated rat model. Arch Ophthalmol. 2001;119(3):399–404.
210. Kim SJ, Toma HS. Inhibition of choroidal neovascularization by intravitreal ketorolac. Arch
Ophthalmol. 2010;128(5):596–600.
211. Olson JL, et al. Intravitreal anakinra inhibits choroidal neovascular membrane growth in a rat model.
Ocul Immunol Inflamm. 2009;17(3):195–200.
212. Rennel ES, et al. A human neutralizing antibody specific to Ang-2 inhibits ocular angiogenesis.
Microcirculation. 2011;18(7):598–607.
213. Olson JL, Courtney RJ, Mandava N. Intravitreal infliximab and choroidal neovascularization in an
animal model. Arch Ophthalmol. 2007;125(9):1221–1224.
214. Zou Y, Xu X, Chiou GC. Effect of interleukin-1 blockers, CK112, and CK116 on rat experimental
choroidal neovascularization in vivo and endothelial cell cultures in vitro. J Ocul Pharmacol Ther.
2006;22(1):19–25.
215. Fu Y, et al. Angiogenesis inhibition and choroidal neovascularization suppression by sustained
delivery of an integrin antagonist, EMD478761. Invest Ophthalmol Vis Sci. 2007;48(11):5184–5190.
216. Pan CK, et al. Comparison of long-acting bevacizumab formulations in the treatment of choroidal
neovascularization in a rat model. J Ocul Pharmacol Ther. 2011;27(3):219–224.
217. Reich SJ, et al. Small interfering RNA (siRNA) targeting VEGF effectively inhibits ocular
neovascularization in a mouse model. Mol Vis. 2003;9:210–216.
218. Huang H, Shen J, Vinores SA. Blockade of VEGFR1 and 2 suppresses pathological angiogenesis and
vascular leakage in the eye. PLoS One. 2011;6(6):e21411.
219. Saishin Y, et al. VEGF-TRAP(R1R2) suppresses choroidal neovascularization and VEGF-induced
breakdown of the blood-retinal barrier. J Cell Physiol. 2003;195(2):241–248.
220. Takahashi K, et al. Intraocular expression of endostatin reduces VEGF-induced retinal vascular
permeability, neovascularization, and retinal detachment. FASEB J;2003;17(8):896–898.
221. Balaggan KS, et al. EIAV vector-mediated delivery of endostatin or angiostatin inhibits angiogenesis
and vascular hyperpermeability in experimental CNV. Gene Ther. 2006;13(15):1153–1165.
222. Lai CC, et al. Suppression of choroidal neovascularization by adeno-associated virus vector
expressing angiostatin. Invest Ophthalmol Vis Sci. 2001;42(10):2401–2407.
223. Mori K, et al. AAV-mediated gene transfer of pigment epithelium-derived factor inhibits choroidal
neovascularization. Invest Ophthalmol Vis Sci. 2002;43(6):1994–2000.
224. Ryan SJ. Subretinal neovascularization after argon laser photocoagulation. Albrecht Von Graefes Arch
Klin Exp Ophthalmol. 1980;215(1):29–42.
225. Ryan SJ. Subretinal neovascularization. Natural history of an experimental model. Arch Ophthalmol.
1982;100(11):1804–1809.
226. Criswell MH, et al. The squirrel monkey: characterization of a new-world primate model of
experimental choroidal neovascularization and comparison with the macaque. Invest Ophthalmol Vis Sci.
2004;45(2):625–634.
227. Ishibashi T, et al. Morphologic observations on experimental subretinal neovascularization in the
monkey. Invest Ophthalmol Vis Sci. 1987;28(7):1116–1130.
228. Miller H, et al. Pathogenesis of laser-induced choroidal subretinal neovascularization. Invest
Ophthalmol Vis Sci. 1990;31(5):899–908.
229. Criswell MH, et al. Anastomotic vessels remain viable after photodynamic therapy in primate models
of choroidal neovascularization. Invest Ophthalmol Vis Sci. 2005;46(6):2168–2174.
230. Ohkuma T, et al. Experimental study on expanded polytetrafluoroethylene (E-PTFE) for portal vein
reconstruction with combined operation of the digestive tract. J Cardiovasc Surg (Torino).
1982;23(4):328–333.
231. Shen WY, et al. Preclinical evaluation of a phosphorothioate oligonucleotide in the retina of rhesus
monkey. Lab Invest. 2002;82(2):167–182.
232. Goody RJ, et al. Optimization of laser-induced choroidal neovascularization in African green
monkeys. Exp Eye Res. 2011;92(6):464–472.
233. Tolentino MJ, et al. Intravitreal injection of vascular endothelial growth factor small interfering RNA
inhibits growth and leakage in a nonhuman primate, laser-induced model of choroidal neovascularization.
Retina. 2004;24(4):660.
234. Zahn G, et al. Preclinical evaluation of the novel small-molecule integrin alpha5beta1 inhibitor
JSM6427 in monkey and rabbit models of choroidal neovascularization. Arch Ophthalmol.
2009;127(10):1329–1335.
235. elDirini AA, Ogden TE, Ryan SJ. Subretinal endophotocoagulation. A new model of subretinal
neovascularization in the rabbit. Retina. 1991;11(2):244–249.
236. Kiilgaard JF, et al. A new animal model of choroidal neovascularization. Acta Ophthalmol Scand.
2005;83(6):697–704.
237. Goldberg MF. Editorial: Bruch's membrane and vascular growth. Invest Ophthalmol.
1976;15(6):443–446.
238. Lassota N, et al. Surgical induction of choroidal neovascularization in a porcine model. Graefes Arch
Clin Exp Ophthalmol. 2007;245(8):1189–1198.
239. Sarks SH. New vessel formation beneath the retinal pigment epithelium in senile eyes. Br J
Ophthalmol. 1973;57(12):951–965.
240. Sarks JP, Sarks SH, Killingsworth MC. Morphology of early choroidal neovascularisation in age-
related macular degeneration: correlation with activity. Eye (Lond). 1997;11(Pt 4):515–522.
241. Zhao L, et al. Translocation of the retinal pigment epithelium and formation of sub-retinal pigment
epithelium deposit induced by subretinal deposit. Mol Vis. 2007;13:873–880.
242. Qiu G, et al. A new model of experimental subretinal neovascularization in the rabbit. Exp Eye Res.
2006;83(1):141–152.
243. Kleinman HK, Martin GR. Matrigel: basement membrane matrix with biological activity. Semin
Cancer Biol. 2005;15(5):378–386.
244. Wang ZY, et al. Erythropoietin as a novel therapeutic agent for atrophic age-related macular
degeneration. Med Hypotheses. 2009;72(4):448–450.
245. Tamai K, et al. Lipid hydroperoxide stimulates subretinal choroidal neovascularization in the rabbit.
Exp Eye Res. 2002;74(2):301–308.
246. Baba T, et al. A rat model for choroidal neovascularization using subretinal lipid hydroperoxide
injection. Am J Pathol. 2010;176(6):3085–3097.
247. Bora PS, et al. Role of complement and complement membrane attack complex in laser-induced
choroidal neovascularization. J Immunol. 2005;174(1):491–497.
248. Lyzogubov VV, et al. Polyethylene glycol (PEG)-induced mouse model of choroidal
neovascularization. J Biol Chem. 2011;286(18):16229–16237.
249. Grossniklaus HE, et al. Macrophage and retinal pigment epithelium expression of angiogenic
cytokines in choroidal neovascularization. Mol Vis. 2002;8:119–126.
250. Oh H, et al. The potential angiogenic role of macrophages in the formation of choroidal neovascular
membranes. Invest Ophthalmol Vis Sci. 1999;40(9):1891–1898.
251. Jo YJ, et al. Establishment of a new animal model of focal subretinal fibrosis that resembles disciform
lesion in advanced age-related macular degeneration. Invest Ophthalmol Vis Sci. 2011;52(9):6089–6095.
252. Blaauwgeers HG, et al. Polarized vascular endothelial growth factor secretion by human retinal
pigment epithelium and localization of vascular endothelial growth factor receptors on the inner
choriocapillaris. Evidence for a trophic paracrine relation. Am J Pathol. 1999;155(2):421–428.
253. Miller JW, et al. Vascular endothelial growth factor/vascular permeability factor is temporally and
spatially correlated with ocular angiogenesis in a primate model. Am J Pathol. 1994;145(3):574–584.
254. Kvanta A, et al. Subfoveal fibrovascular membranes in age-related macular degeneration express
vascular endothelial growth factor. Invest Ophthalmol Vis Sci. 1996;37(9):1929–1934.
255. Lopez PF, et al. 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. 1996;37(5):855–868.
256. Okamoto N, et al. Transgenic mice with increased expression of vascular endothelial growth factor in
the retina: a new model of intraretinal and subretinal neovascularization. Am J Pathol.
1997;151(1):281–291.
257. Kwak N, et al. VEGF is major stimulator in model of choroidal neovascularization. Invest Ophthalmol
Vis Sci. 2000;41(10):3158–3164.
258. Takeda A, et al. CCR3 is a target for age-related macular degeneration diagnosis and therapy. Nature.
2009;460(7252):225–230.
259. Stewart MW. The expanding role of vascular endothelial growth factor inhibitors in ophthalmology.
Mayo Clin Proc. 2012;87(1):77–88.
260. Meyer CH, Holz FG. Preclinical aspects of anti-VEGF agents for the treatment of wet AMD:
ranibizumab and bevacizumab. Eye (Lond). 2011;25(6):661–672.
261. Bakri SJ, et al. Pharmacokinetics of intravitreal bevacizumab (Avastin). Ophthalmology.
2007;114(5):855–859.
262. Bakri SJ, et al. Pharmacokinetics of intravitreal ranibizumab (Lucentis). Ophthalmology.
2007;114(12):2179–2182.
263. Gaudreault J, et al. Preclinical pharmacokinetics of Ranibizumab (rhuFabV2) after a single intravitreal
administration. Invest Ophthalmol Vis Sci. 2005;46(2):726–733.
264. Thaler S, et al. Toxicity testing of the VEGF inhibitors bevacizumab, ranibizumab and pegaptanib in
rats both with and without prior retinal ganglion cell damage. Acta Ophthalmol. 2010;88(5):e170–e176.
265. Miki K, et al. Effects of intraocular ranibizumab and bevacizumab in transgenic mice expressing
human vascular endothelial growth factor. Ophthalmology. 2009;116(9):1748–1754.
266. Maier P, et al. Intravitreal injection of specific receptor tyrosine kinase inhibitor PTK787/ZK222 584
improves ischemia-induced retinopathy in mice. Graefes Arch Clin Exp Ophthalmol. 2005;243(6):593–600.
267. Thumann G. Prospectives for gene therapy of retinal degenerations. Curr Genomics.
2012;13(5):350–362.
268. Chizzolini M, et al. Good epidemiologic practice in retinitis pigmentosa: from phenotyping to
biobanking. Curr Genomics. 2011;12(4):260–266.
269. Inglehearn CF, et al. A linkage survey of 20 dominant retinitis pigmentosa families: frequencies of the
nine known loci and evidence for further heterogeneity. J Med Genet. 1998;35(1):1–5.
270. Wang DY, et al. Gene mutations in retinitis pigmentosa and their clinical implications. Clin Chim
Acta. 2005;351(1–2):5–16.
271. Thompson DA, et al. Rd9 is a naturally occurring mouse model of a common form of retinitis
pigmentosa caused by mutations in RPGR-ORF15. PLoS One. 2012;7(5):e35865.
272. Ross JW, et al. Generation of an inbred miniature pig model of retinitis pigmentosa. Invest
Ophthalmol Vis Sci. 2012;53(1):501–507.
273. Aguirre G. Retinal degenerations in the dog. I. Rod dysplasia. Exp Eye Res. 1978;26(3):233–253.
274. May CA, Lutjen-Drecoll E, Narfstrom K. Morphological changes in the anterior segment of the
Abyssinian cat eye with hereditary rod-cone degeneration. Curr Eye Res. 2005;30(10):855–862.
275. Cheung N, Mitchell P, Wong TY. Diabetic retinopathy. Lancet. 2010;376(9735):124–136.
276. Robinson R, et al. Update on animal models of diabetic retinopathy: from molecular approaches to
mice and higher mammals. Dis Model Mech. 2012;5(4):444–456.
277. Kern TS, et al. Comparison of three strains of diabetic rats with respect to the rate at which
retinopathy and tactile allodynia develop. Mol Vis. 2010;16:1629–1639.
278. Sima AA, et al. The BB-rat—an authentic model of human diabetic retinopathy. Curr Eye Res.
1985;4(10):1087–1092.
279. Danis RP, Yang Y. Microvascular retinopathy in the Zucker diabetic fatty rat. Invest Ophthalmol Vis
Sci. 1993;34(7):2367–2371.
280. Matsuura T, et al. Proliferative retinal changes in diabetic rats (WBN/Kob). Lab Anim Sci.
1999;49(5):565–569.
281. Miyamura N, Amemiya T. Lens and retinal changes in the WBN/Kob rat (spontaneously diabetic
strain). Electron-microscopic study. Ophthalmic Res. 1998;30(4):221–232.
282. Goto Y, et al. Development of diabetes in the non-obese NIDDM rat (GK rat). Adv Exp Med Biol.
1988;246:29–31.
283. Yamada H, et al. Retinal neovascularisation without ischaemia in the spontaneously diabetic Torii rat.
Diabetologia. 2005;48(8):1663–1668.
284. Makino S, et al. Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu. 1980;29(1):1–13.
285. Midena E, et al. Studies on the retina of the diabetic db/db mouse. I. Endothelial cell-pericyte ratio.
Ophthalmic Res. 1989;21(2):106–111.
286. Barber AJ, et al. The Ins2Akita mouse as a model of early retinal complications in diabetes. Invest
Ophthalmol Vis Sci. 2005;46(6):2210–2218.
287. Johnson MA, et al. Ocular structure and function in an aged monkey with spontaneous diabetes
mellitus. Exp Eye Res. 2005;80(1):37–42.
3
PATHOGENESIS AND
PATHOPHYSIOLOGY OF AGE-
RELATED MACULAR
DEGENERATION ANA
ANA MACHÍN MAHAVE • ERNESTO ROMERA REDONDO
JOHN BARNWELL KERRISON
INTRODUCTION
Age-related macular degeneration (AMD) is the leading cause of severe visual
acuity loss in the United States among people older than 60 years, representing
54% of legal blindness (1). In the general population, the number of people in
this group is increasing. As a result, vision loss from macular degeneration is a
growing problem. Ninety percent of AMD patients have dry AMD, and 10% of
AMD patients have wet AMD. Currently, an estimated 8 million Americans are
affected with early AMD, and over 1 million will develop advanced AMD
within the next 5 years (2).
Many changes in the macula could be due to normal aging. The focus of
study has been which aging processes are implicated in the pathogenesis of
senile macular degeneration or at what stage they become pathologic (3). The
critical question to be answered is “Is AMD a normal process of aging or is it a
disease by itself?” (4–7). Most of the authors defend the theory in which AMD is
an advanced stage or perturbation of the normal process of eye senescence
(3,4,8,9).
The pathophysiology of AMD is incompletely understood, but genetic tools
offer new insights into the development and progress of AMD (10).
RISK FACTORS
AMD is a multifactorial disease in which multiple genetic and environmental
factors are involved.
Age: Age is the largest risk factor for AMD (11–17).
Smoking: The mechanism by which smoking might affect the retina is
unknown; however, there exists a direct association between the risk of
developing advanced AMD with the number of cigarettes smoked (18,19).
Sunlight exposure: Individuals who wore sunglasses regularly were less
likely to develop soft drusen (20). Results from the Beaver Dam Study suggest
that people who spent leisure time outdoors were at an increased risk of
developing early AMD (21). On the other hand, some studies have shown little
or no association between sunlight exposure and the risk of AMD (22–24).
White individuals: It is postulated that increased levels of melanin could
increase the free radical scavenging potential of the retinal pigment epithelium
(RPE) and Bruch's membrane (BrM), thereby protecting against the risk of
advanced AMD (25–28).
Female sex: Female gender might be a risk factor in individuals older than
75 years (29), with double the risk of developing wet AMD in comparison with
age-matched men. However, nonstatistically significant differences have been
demonstrated (30).
Cholesterol levels, obesity, hypertension, cardiovascular disease, and
increased dietary fat: These factors seem to be related to the risk of developing
AMD, but different studies do not agree (31–37).
Genetics: Several studies show a genetic factor in the pathogenesis of the
disease (38–41). The gene for apolipoprotein E (APOE) is the first identified
susceptibility gene for AMD and has been also associated with other diseases of
aging including Alzheimer's disease. APOE epsilon 2 allele is associated with a
50% increased risk of AMD (42); however, APOE epsilon 4 allele is associated
with 57% reduction in risk of wet AMD (43). Several loci have been also
associated with AMD, including two major loci in the complement factor H
(CFH) gene on 1q32 and the ARMS2/HTRA1 locus on the 10q26 gene cluster
(44,45).
In short, genes influence many biologic pathways, but genetic susceptibility
can be modified by environmental factors. Together, they are greatly predictive
of onset and progression of disease (46,47).
TRANSPORT
Transport through the RPE is bidirectional. From the subretinal space to choroid,
the RPE transports electrolytes and water. From the blood to the photoreceptors,
the RPE transports glucose and other nutrients.
Transport from blood to the photoreceptors encompasses glucose and other
vital nutrients. The RPE absorbs glucose, retinol, ascorbic acid, and fatty acids in
the blood and delivers them to the photoreceptor. For the transport of glucose,
the RPE has high amounts of glucose transporter (GLUT) in both their apical
and basolateral membranes. Another notable function of the RPE is transport of
retinyl to ensure its supply to the retinal photoreceptors. In this process, many
complex intermediate steps take place. A critical step involves an RPE enzyme
that isomerizes all-trans-retinyl esters into 11-cis-retinal, which is essential for
rod and cone function (33,53).
The delivery of the fatty acids such as docosahexaenoic acid and
eicosapentaenoic acid to the photoreceptor is another important RPE function
(54). These omega-3 fatty acids cannot be synthesized by the nervous tissue but
are an essential component of the membranes of neurons and photoreceptors.
They are synthesized in the liver from the precursor, linolenic acid, and are
carried in the blood by plasma lipoproteins (33). In addition, docosahexaenoic
acid is the precursor of the D1 neuroprotectin that protects RPE from oxidative
stress (25,55). The main sources of omega-3 fatty acid are fish products. The
Blue Mountain Eye Study and some case–control studies have found evidence of
reduced risk of advanced AMD and in those who eat fish regularly (34,56,57).
Transport from photoreceptors to blood is important for ions and water. The
RPE transports ions and water from the subretinal space (apical side) into the
blood of the choroid (basolateral side). This process requires energy, and the Na
(+)/K (+)-ATPase, on the apical membrane of the RPE, provides the energy (58).
Tight junctions between RPE cells result in a barrier between the subretinal
space and the choriocapillaris. Notably, paracellular resistance of this barrier is
10 times the transcellular resistance. For this reason, water cannot pass through
the paracellular RPE pathway and passes mainly through the transcellular
pathway via aquaporin-1 (59,60).
PHAGOCYTOSIS
The RPE is a phagocytic system that is essential for the renewal of
photoreceptors. Each photoreceptor has an inner and outer segment, the outer
segment of each cone has a membrane folded 700 times, and the outer segment
of every rod has about 1,000 discs (62).
The photoreceptors are often exposed to constant high light levels, which
leads to accumulation of oxidized proteins and lipids. The concentration of
photo-oxidized substances increases within the photoreceptors daily. The
transduction of the light depends on the photoreceptor by appropriate
functioning and structure of the proteins, retinyl, and cell membranes. Therefore,
to maintain the excitability of the photoreceptor outer segments, they are
undergoing constant renewal, reconstructed from its base. The tips of the outer
segments of the photoreceptors, containing the highest concentration of free
radicals, proteins, and photo-oxidized lipids, detach from the photoreceptors.
Outer segments maintain a constant length through the coordinated release of its
extremities and the formation of new discs. The discs detach from the outer
segments and are phagocytosed by the RPE (63), which digest and deliver the
essential molecules, such as docosahexaenoic acid and retinyl, back to the
photoreceptors to rebuild their light-sensitive outer segments (32,64). Sometimes
the content of the phagolysosomes are incompletely degraded, forming the
residual bodies that are the substrates for lipofuscin formation. The residual
bodies in the RPE accumulate with age, beginning as early as 16 months (65,66).
The limited autophagocytic capacity of the RPE eventually results in an
accumulation of metabolic waste in RPE cells throughout life. Dry AMD often
begins in the parafoveal ring, where lipofuscin concentration is highest.
However, lipofuscin disappears with cell death and a reduction in
autofluorescent lipofuscin is a sign of progression toward late AMD.
SECRETION
RPE cells produce and secrete growth factors as well as essential factors for
maintaining structural integrity of the retina and choroid. These molecules favor
the photoreceptors' survival and ensure a structural integrity for optimal
circulation and nutrient supply. RPE secretes pigment epithelium–derived factor
(PEDF), vascular endothelial growth factor (VEGF), fibroblast growth factors
(FGF-1, FGF-2, and FGF-5), transforming growth factor–β, insulin-like growth
factor I, neuronal growth factor (NGF), growth factor derived from the brain,
neurotrophin-3, ciliary neurotrophic factor, several interleukins, chemokines,
tumor necrosis factor–α, colony-stimulating factors, and various types of
metalloprotease tissue inhibitors (67,68).
The most important factors with regard to the pathophysiology of AMD are
PEDF and VEGF. PEDF is a neuroprotection and antiangiogenic factor. PEDF
inhibits endothelial cell proliferation and stabilizes choriocapillaris endothelium.
It is important in embryonic eye development. Mice lacking PEDF develop more
aggressive retinal vascularization in ischemia (69). VEGF is secreted in low
concentrations during physiologic conditions, thereby preventing endothelial cell
apoptosis. In addition, VEGF regulates vascular permeability, thus stabilizing
endothelium fenestrations (70).
IMMUNE PRIVILEGE
The eye is an immune-privileged site, and the RPE cells have an important role
in the maintenance of this immune privilege (71,72). In 1993, Liversidge et al.
indicated how RPE cells could inhibit lymphocyte proliferation (73). Many
pathways have been suggested to explain RPE immunosuppression (74). On the
one hand, RPE inhibits T-cell proliferation through secretion of prostaglandins
and immunosuppressive cytokines. On the other hand, apoptosis and
phagocytosis properties have demonstrated both directly (decreasing T-cell
number and clearing apoptotic lymphocytes) and indirectly (down-regulating
proinflammatory cytokine secretion, such as IL-1b) (75,76).
Ambati et al. (77) in 2003 postulated in a study of mice that there is an
accumulation of complement fragments that might damage RPE and induce
VEGF production by RPE cells, resulting in the development of choroidal
neovascularization (CNV). This study provides insight into the role of
macrophages and complement in the pathophysiology of AMD (78).
BRUCH'S MEMBRANE
BrM is a complex pentalaminar extracellular matrix (ECM), located beneath the
RPE, and includes three layers: a central elastic layer sandwiched between two
collagenous layers. BrM is lined by the basement membranes of the RPE and the
choriocapillaris (basal laminae). Apart from its structural support function, BrM
is also highly permeable to fluid and small molecules like oxygen, glucose, and
other molecules necessary to maintain retinal health and function (79).
Proteoglycans play an important role in BrM (80). Their negative charge impairs
the passage of positively charged macromolecules that are essential for
maintenance of RPE.
Changes in BrM begin in the third or fourth decade of life (5,81,82). These
changes include the accumulation of membranous debris on both sides of the
elastic lamina, drusen between RPE basal lamina and inner collagenous layer of
BrM, and basal laminar deposits that may occupy the same space as drusen or
develop between the RPE cell and its basal lamina (79) and appears as an
amorphous material. How drusen develop and why they can vary in location and
features are unknown.
Drusen usually contain fragments of RPE cells (9), a core of glycoproteins,
an outer dome of crystallins (83,84), chaperone proteins, APOE, vitronectin, and
proteins related to inflammation (amyloid P, C5, and C5b–9 complement
complex) (85). Drusen are classified as either hard or soft. Hard drusen look like
small, yellow nodules and are not associated with AMD (86). Soft drusen appear
as large, white or light yellow, dome-shaped elevations that can resemble
localized serous RPE detachments.
BrM doubles in thickness and declines in elasticity between the ages of 10
and 90 years. It contains no lipids during the first 30 years of life, but lipid
concentrations rise in later years (4). In addition to the structural alterations that
take place in BrM during aging, its fluid permeability becomes substantially
reduced. BrM acts as a scaffold for the choriocapillaris and RPE cells, regulating
their survival. When this function is diminished, anoikis occurs. Anoikis is an
apoptosis phenomenon resulting from incorrect cell adhesion, and this process
takes place in photoreceptors, RPE cells, and possibly endothelial cells (4) of the
choriocapillaris. The development of either excessive material in BrM or erosion
of BrM has important consequences. It may lead to choroidal endothelial cell
loss, instigate chronic local inflammation, and produce ischemic stress on the
oxygen hungry photoreceptor cells.
CHORIOCAPILLARIS
Choriocapillaris is a plexus of capillaries with fenestrations on the side facing
BrM and is part of the innermost layer in the choroid. The capillary network is
designed in a lobular pattern for rapid blood flow to supply the high metabolic
demand of the photoreceptors and RPE (82). In aging eyes, the lumina of the
choriocapillaris and the choroidal thickness are reduced by half. The resulting
hypoxia stabilizes hypoxia-inducible factor 1α, which activates genes encoding
proteins such as erythropoietin that protect the photoreceptors (87). Hypoxia also
increases the secretion of growth factors such as VEGF A within Ruysch's
complex on the basal side of the RPE cells, causing development of choroidal
neovascular membranes (4,88).
GEOGRAPHIC ATROPHY
Deposits of cellular debris in BrM alter the pathway of nutrient exchange
between the RPE and choriocapillaris and lead to the downturn of RPE cells. At
the same time, RPE cells overlying drusen are usually inflated and depigmented,
and the adjacent photoreceptors are often damaged or dead (8). When the RPE
degeneration is more advanced, thick basal laminar deposits take place in the
BrM and obstruct nutrient exchange. These deposits may also decrease the
adhesion of the RPE to the inner collagenous layer of BrM facilitating serous
detachment of the RPE and retina (89,90).
Throughout their history, drusen reflect the state of the adjacent RPE cells.
The greater the number of drusen and the larger they are, the greater the
probability that the RPE cells will be damaged (8). Soft drusen easily became
confluent and can reach a size that produces serous detachments of the RPE (91).
Drusen sometimes disappear. Macrophages, pericytes, and choriocapillaris
contribute to the removal, leading to their replacement by fibrous tissue.
By this time, RPE deterioration is critically advanced, and some cells may
proliferate into small clumps, detach from BrM, and migrate into the retina. The
final consequence of this process is geographic atrophy. Depigmentation exists
in atrophic areas; however, at the edge of the lesions, pigmentation develops due
to proliferation, hypertrophy, or phagocytosis of liberated melanin and lipofuscin
(8,92). During the process of geographic atrophy, neural retina and damaged
RPE cells disappear and the surviving rods and cones become shorter.
DISCIFORM SCAR
The main site of disturbance in neovascular AMD is BrM. However, the changes
in BrM that predispose to neovascularization are unclear. With aging and as a
consequence of membrane calcification, small fractures or gaps develop in BrM.
Abnormal secretion from the deteriorating RPE cells might serve as attractant
for endothelial cells, phagocytic cells, and lymphocytes, resulting in
neovascularization. Lipofuscin accumulation, RPE membrane thickening, and
intracellular residual body increase help stimulate new vessel formation.
Therefore, new vessels occur as a consequence of an imbalance in the
stimulating and inhibiting influences of growth factors, and any disruption in the
diffusion through BrM to choroid could alter this balance. Once established, the
new vessels have tendency to leak and bleed. When this occurs, the fluid extends
laterally beneath the RPE in the same plane where basal laminar deposit and
soften drusen are located.
There are three types of pigment epithelial detachment (PED) associated
with AMD: drusenoid, serous, and fibrovascular. Drusenoid PED is a confluence
of several soft drusen. Fibrovascular PED is a form of occult CNV. Serous PED
is not exactly a breakdown in the outer BRB, it seems to develop from a
dysfunction in the complex RPE cells–BrM. Fluid moves from the retina into the
BrM as a result of active movement of ions by the RPE cells. If BrM became
hydrophobic, resistance to water flow could cause fluid to collect between RPE
and BrM (74). Serous exudates and blood may enter the retinal space. The
retinal detachment may extend beyond the zone of RPE detachment and result in
a rapid destruction of the RPE and photoreceptors over an area much larger than
the region of vascular invasion, so the loss of vision is more severe in
neovascular AMD than it is in geographic atrophy. Hemorrhage under the RPE
or retina stimulates proliferation of fibrous tissue that produces a disciform scar,
increasing the size of the area of blindness.
CONCLUSION
The main features of the AMD are the formation of drusen; thickening and other
signs of damage of BrM; depigmentation and RPE hyperplasia; and
accumulation of basal laminar deposits beneath the RPE. However, these are not
unique or pathognomonic of this disease as we often observe them in most eyes
of elderly individuals (93–96). In summary, the main signs of AMD increase
with age, but only in some persons, they progress to the stage of cell death and
functional loss. According to this point of view, AMD is an advanced stage of a
process that occurs in all eyes. It is part of a common and continuing process in
which the transition between age and disease is marked by loss of vision (9).
In 1972, Hogan (97) described the role RPE plays in the macula and
postulated the existence of a gradual decrease in the activity of RPE, which
resulted in accumulation of metabolic waste in BrM and loss of permeability.
Zarbin (10) summarized five key concepts:
REFERENCES
1. Congdon N, O'Colmain B, Klaver CC, et al. Causes and prevalence of visual impairment among adults
in the United States. Arch Ophthalmol. 2004;122:477–485.
2. Age-related Eye Disease Study Research Group. Potential public health impact of age-related disease
study results. Arch Ophthalmol. 2003;121:1621–1624.
3. Sharks SH. Ageing and degeneration in the macular region: a clinic-pathological study. Brit J
Ophthalmol. 1976;60:324–341.
4. de Jong PT. Mechanisms of disease. N Engl J Med. 2006;355:1474–1485.
5. Hogan MJ, Alvarado J. Studies on the human macula. IV. Aging changes in Bruch's membrane. Arch
Ophthalmol. 1967;77:410–420.
6. Peto R, Doll R. There is no such thing as aging. BMJ. 1997;315:1030–1032.
7. Powell DE. There is no such thing as ageing: ageing has been defined as to grow or make old. BMJ.
1998;316:1531.
8. Young RW. Pathophysiology of age-related macular degeneration. Surv Ophthalmol.
1987;31(5):291–306.
9. Zarbin MA. Current concept in the pathogenesis of age-related macular degeneration. Arch Ophthalmol.
2004;122:598–614.
10. Handa JT. New molecular histopathologic insights into the pathogenesis of age-related macular
degeneration. Int Ophthalmol Clin. 2007;47(1):15–50.
11. Bird AC, Bressler NM, Bressler SB, et al. The International ARM Epidemiological Study Group. An
international classification and grading system for age-related maculopathy and age related macular
degeneration. Surv Ophthalmol. 1995;39:367–374.
12. Mitchell P, Smith W, Attebo K, et al. Prevalence of age-related maculopathy in Australia.
Ophthalmology. 1995;102:1450–1460.
13. Klein R, Klein BE, Linton KL. Prevalence of age-related maculopathy. Ophthalmology.
1992;99:933–943.
14. Cruickshanks KJ, Hammar RF, Klein R, et al. The prevalence of age-related maculopathy by
geographic region and ethnicity. Arch Ophthalmol. 1997;115:242–250.
15. Friedman DS, Katz J, Bressler NM, et al. Racial differences in the prevalence of age-related macular
degeneration: the Baltimore eye survey. Ophthalmology. 1999;106:1049–1055.
16. Bressler NM, Bressler SB, West SK, et al. The grading and prevalence of macular degeneration in
Chesapeake bay waterman. Arch Ophthalmol. 1989;107:847–852.
17. Van Newkirk MR, Nanjan MB, Wanh JJ, et al. The prevalence of age-related maculopathy: the visual
impairment project. Ophthalmology. 2000;107: 1593–1600.
18. Delcourt C, Diaz JL, Ponton-Sanchez A, et al. Smoking and age-related macular degeneration. Arch
Ophthalmol. 1998;116:1031–1035.
19. Khan JC, Thurlby DA, Shahid H, et al. Smoking and age-related macular degeneration: the number of
pack years of cigarette smoking is a major determinant of risk for both geographic atrophy and choroidal
neovascularization. Br J Ophthalmol. 2006;90:75–80.
20. Delcourt C, Carriere I, Ponton-Sanchez A, et al. Light exposure and the risk of age-related macular
degeneration: the POLA Study. Arch Ophthalmol. 2001;119:1463–1468.
21. Klein R, Klein BE, Jensen SC, et al. Sunlight and the 5-year incidence of early age-related
maculopathy: the Beaver Dam Eye Study. Arch Ophthalmol. 2001;119:246–250.
22. The Age-Related Eye Disease Study Research Group. Risk factors associated with age-related macular
degeneration: a case control study in the age-related eye disease study. Ophthalmology.
2000;107:2224–2232.
23. The Eye Disease Case-control Study Group. Risk factors for neovascular age-related macular
degeneration. Arch Ophthalmol. 1992;110:1701–1708.
24. Klein R. Epidemiology. In: Berger JW, Fine SL, Maguire MG, eds. Age-related macular degeneration.
St. Louis, MO: Mosby; 1999:31–55.
25. Coleman HR, Chan CH, Ferris III FL, et al. Age-related macular degeneration. Lancet.
2008;372:1835–1845.
26. Mukesh BN, Dimitrov PN, Leikin S, et al. Five year incidence of age-related maculopathy: visual
impairment project. Ophthalmology. 2004;111:1176–1182.
27. Klein R, Klein BE, Jenson SC, et al. Age-related maculopathy in a multiracial United States
population: the National Health and Nutrition Examination Survey III. Ophthalmology.
1999;106:1056–1065.
28. Schachat AP, Hyman L, Leske MC, et al. Features of age-related macular degeneration in a black
population. Arch Ophthalmol. 1995;113:728–735.
29. Smith W, Assink J, Klein R, et al. Risk factors for age-related macular degeneration: pooled findings
from three continents. Ophthalmology. 2001;108:697–704.
30. Mitchell P, Wang JJ, Foran S, et al. Five-year incidence of age-related maculopathy lesions: the Blue
Mountain Eye Study. Ophthalmology. 2002;109:1092–1097.
31. Mares-Perlman JA, Brady WE, et al. Dietary fat and age-related maculopathy. Arch Ophthalmol.
1995;113:743–748.
32. Cho E, Hung S, Willet C, et al. Prospective study of dietary fat and the risk of age-related macular
degeneration. Am J Clin Nutr. 2001;73:209–218.
33. Heuberger RA, Mares-Pearlman JA, Klein R, et al. Relationship of dietary fat to age-related
maculopathy in the Third National Health and Nutrition Examination Survey. Arch Ophthalmol.
2001;119:1833–1838.
34. Seddon JM, Rosner B, Sperduto RD, et al. Dietary fat and risk for advanced age-related macular
degeneration. Arch Ophthalmol. 2001;119:1191–1199.
35. Klein R, Klein BE, Tomany SC, et al. The association of cardiovascular disease with the long-term
incidence of age-related maculopathy. Ophthalmology. 2003;110:1273–1280.
36. Fraser-Bell S, Wu J, Klein R, et al. Cardiovascular risk factors and age-related macular degeneration:
the Los Angeles Latino Eye Study. Am J Ophthalmol. 2008;145:308–316.
37. Hogg RE, Woodside JV, Gilchrist SE, et al. Cardiovascular disease and hypertension are strong risk
factors for choroidal neovascularization. Ophthalmology. 2008;115:1046–1052.
38. Traboulsi Ei. The challenges and surprises of studying the genetics of age-related macular
degeneration. Am J Ophthalmol. 2005;139:908–911.
39. Piguet B, Wells JA, Palmvang IB, et al. Age-related Bruch's membrane change: a clinical study of the
relative role of heredity and environment. Br J Ophthalmol. 1993;77:400–403.
40. Hamdi HK, Kenney C. Age-related macular degeneration: a new viewpoint, Front Biosci.
2003;8:305–314.
41. Zareparsi S, Reddick AC, Branham KE, et al. Association of apolipoprotein E alleles with
susceptibility to age-related macular degeneration in a large cohort from a single center. Invest Ophthalmol
Vis Sci. 2004;45:1306–1310.
42. Klaver CC, Kliffen M, Van Duijn CM, et al. Genetic association of apolipoprotein E with age-related
macular degeneration. Am J Hum Genet. 1998;63:200–206.
43. Souied EH, Benlian P, Amouyel P, et al. The epsilon 4 allele of the apolipoprotein E gene as a potential
protective factor of exudative age-related macular degeneration. Am J Ophthalmol. 1998;125:353–359.
44. Allikmets R. Simple and complex ABCR: genetic predisposition to retinal disease. Am J Hum Genet.
2000;67:793–799.
45. Weeks De, Conley YP, Tsai HJ, et al. Age-related maculopathy: a genomewide scan with continued
evidence of susceptibility loci within the 1q31, 10q26 and 17q25 regions. Am J Hum Genet.
2004;75(2):174–189.
46. Lim LS, Mitchell P, Seddon JM, et al. Age-related macular degeneration. Lancet.
2012;379:1728–1738.
47. Seddon JM, Reynolds R, Yu Y, et al. Risk models for progression to advanced age-related macular
degeneration using demographic, environmental, genetic, and ocular factors. Ophthalmology.
2011;118:2203–2211.
48. Ruysch F. Asser primus: thesaurus anatomicus secundus. Amsterdam, The Netherlands: J.Waesbergios;
1722:3–14.
49. Green WR. Histopathology of age-related macular degeneration. Mol Vis. 1999;5:27.
50. Green WR, Enger C. Age-related macular degeneration histopathologic studies. The 1992 Lorenz E
Zimmerman Lecture. Ophthalmology. 1993;100:1519–1535.
51. Thebault S. El epitelio pigmentario retiniano como componente de la barrera hematoretiniana:
implicación en la retinopatía diabética. Revista Digital Universitaria [en línea]. 1 de marzo de. 2011;12(3).
[Consultada: 2 de marzo de 2011]. Disponible en Internet:
http://www.revista.unam.mx/vol.12/num3/art31/index.html. ISSN: 1607–6079.
52. Strauss O. The retinal pigment epithelium in visual function. Physiol Rev. 2005;85(3):845–881.
53. Redmon TM, Poliakov E, Yu S, et al. Mutation of key residues of RPE65 abolishes its enzymatic role
as isomerohydrolase in the visual cycle. Proc Natl Acad Sci U S A. 2005;102:13658–13663.
54. Bazan NG, Gordon WC, Rodriguez de Turco EB. Docosahexaenoic acid uptake and metabolism in
photoreceptors: retinal conservation by an efficient retinal pigment epithelial cell-mediated recycling
process. Adv Exp Med Biol. 1992;318:295–306.
55. Bazan NG. Neurotrophins induce neuroprotective signaling in the retinal pigment epithelial cell by
activating the synthesis of the anti-inflammatory and anti-apoptotic neuroprotectin D1. Adv Exp Med Biol.
2008;613:39–44.
56. SanGiovanni JP, Chew EY, Clemons TE, et al. The relationship of dietary lipid intake and age-related
macular degeneration in a case–control study. Arch Ophthalmol. 2007;125:671–679.
57. Chua B, Flood V, Rochtchina E, et al. Dietary fatty acids and the 5-year incidence of age-related
maculopathy. Arch Ophthalmol. 2006;124:981–986.
58. Marmorstein AD. The polarity of the retinal pigment epithelium. Traffic. 2001;2(12):867–872.
59. Erickson KK, Sundstrom JM, Antonetti DA. Vascular permeability in ocular disease and the role of
tight junctions. Angiogenesis. 2007;10(2):103–117.
60. Verkman AS, Ruiz-Ederra J, Levin MH. Functions of aquaporins in the eye. Prog Retin Eye Res.
2008;27(4):420–433.
61. Girotti AW, Kriska T. Role of lipid hydroperoxides in photo-oxidative stress signaling. Antioxid Redox
Signal. 2004;6(2):301–310.
62. Young RW. Shedding of discs from rod outer segments in the rhesus monkey. J Ultrastruct Res.
1971;34:190–203.
63. Young RW, Bok D. Participation of the retinal pigment epithelium in the rod outer segment renewal
process. J Cell Biol. 1969;42:392–403.
64. Liu BF, et al. Low phagocytic activity of resident peritoneal macrophages in diabetic mice: relevance to
the formation of advanced glycation end products. Diabetes. 1999;48(10):2074–2082.
65. Streeten BW. The sudanophilic granules of the human retinal pigment epithelium. Arch Ophthalmol.
1961;66:391–398.
66. Wing GL, Blanchard GC, Weiter JJ. The topography and age relationship of lipofuscin concentration in
the retinal pigment epithelium. Invest Ophthalmol Vis Sci. 1978;17:601–607.
67. Witmer AN, et al. Vascular endothelial growth factors and angiogenesis in eye disease. Prog Retin Eye
Res. 2003;22(1):1–29.
68. Stemfeld MD, et al. Cultured human retinal pigment epithelial cells express basic fibroblast growth
factor and its receptor. Curr Eye Res. 1989;8(10):1029–1037.
69. Huang Q, et al. PEDF-deficient mice exhibit and enhanced rate of retinal vascular expansion and are
more sensitive to hyperoxia-mediated vessel obliteration. Exp Eye Res. 2008;87(3):226–241.
70. Lu M, et al. Advanced glycation end products increase retinal vascular endothelial growth factor
expression. J Clin Invest. 1998;101(6):1219–1224.
71. Streilein JW. Peripheral tolerance induction: lessons from immune privileges sites and tissues.
Transplant Proc. 1996;28:2066–2070.
72. Wenkel H, Sreilein JW. Analysis of immune deviation elicited by antigens injected into the subretinal
space. Invest Ophthalmol Vis Sci. 1998;39:1823–1834.
73. Liversidge J, McKay D, Mullen G, et al. Retinal pigment epithelial cells modulate lymphocyte function
at the blood-retina barrier by autocrine PGE2 and membrane-bound mechanisms. Cell Immunol.
1993;149:315–330.
74. Carnota P, Bueno R, Rodriguez-Fontal M, et al. Pathophysiology of the blood-retinal barrier. In:
Quiroz-Mercado H, ed. Macular surgery. 2nd ed. Philadelphia, PA: Lippincott Williams & Wilkins;
2011:17–28.
75. Willermain F, Caspers-Velu L, Nowak B, et al. Retinal pigment epithelial cell phagocytosis of T
lymphocytes: possible implication in the immune privilege of the eye. Br J Ophthalmol.
2002;86:1417–1421.
76. Jorgensen A, Wiencke A, la Cour M, et al. Human retinal pigment epithelial cell-induced apoptosis in
activated T cells. Invest Ophthalmol Vis Sci. 1998;39:1590–1599.
77. Ambati J, et al. An animal model of age related macular degeneration in senescence Ccl-2 or Ccr-2
deficient mice. Nat Med. 2003;9:1390–1397.
78. Tezel TH, Bora NS, Kaplan HJ. Pathogenesis of age-related macular degeneration. Trends Mol Med.
2004;10(9):417–420.
79. Mullins RF, Sohn EH. Bruch's membrane: the critical boundary in macular degeneration. In: Dr. Gui-
Shuang Ying, ed. Degeneration, age related macular degeneration—the recent advances in basic research
and clinical care. InTech; 2012. http://www.intechopen.com/books/age-related-macular-degeneration-the-
recent-advances-in-basic-researchand-clinical-care/bruch-s-membrane-the-critical-boundary-in-amd. ISBN:
978-953-307-864-9.
80. Kliffen M, de Long PT, Luider TM. Protein analysis of human macula in relation to age-related
maculopathy. Lab Invest. 1995;73:262–272.
81. Sarks SH. New vessels formation beneath the retinal pigment epithelium in senile eyes. Br J
Ophthalmol. 1973;57:951–965.
82. Handa JT. Pathophysiology of the retinal pigmented epithelium and Choroid. In: Quiroz-Mercado H,
ed. Macular surgery. 2nd Ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2011:10–16.
83. Crabb JW, Miyagi M, Gu X, et al. Drusen proteome analysis: an approach to the etiology of age-related
macular degeneration. Proc Natl Acad Sci U S A. 2002;99:14682–14687.
84. Hageman GS, Luther PJ, Victor Chong NH, et al. An integrated hypothesis that considers drusen as
biomarkers of immunomediated processes at the RPE-Bruch's membrane interface in aging and age-related
macular degeneration. Prog Retin Eye Res. 2001;20:705–732.
85. Umeda S, Suzuki MT, Okamoto H, et al. Molecular composition of drusen and possible involvement of
anti-retinalautoimmunity in two different forms of macular degeneration in cynomolgus monkey (Macaca
fascicularis). FASEB J. 2005;19:2088.
86. Geen WR, Enger C. Age-related macular degeneration histopathologic studies. The 1992 Lorenz E.
Zimmerman lecture. Ophthalmology. 1993;100:1519–1535.
87. Grimm C, Wenzel A, Groszer M, et al. HIF-1-induced erythropoietin in the hypoxic retina protects
against light induced retinal degeneration. Nat Med. 2002;8:718–724.
88. Dámore PA. Mechanisms of retinal and choroidal neovascularization. Invest Ophthalmol Vis Sci.
1994;35:3974–3979.
89. Braunstein RA, Gass JDM. Serous detachments of the retinal pigment epithelium in patients with senile
macular disease. Am J Ophthalmol. 1979;88:652–660.
90. Gass JDM: Pathogenesis of disciform detachment of the neuroepithelium. III. Senile disciform macular
degeneration. Am J Ophthalmol. 1967;63:617–644.
91. Green WR, Mc Donnell PJ, Yeo JH. Pathologic features of senile macular degeneration.
Ophthalmology. 1985;92:615–627.
92. Burns RP, Feeney-Burns L. Clinico-morphologic correlations of drusen of Bruch´s membrane. Trans
Am Ophthalmol Soc. 1980;78:206-223.
93. Feeney-Burns L, Eldred GE. Age-related changes in the ultrastructure of Bruch´s membrane. Am J
Ophthalmol. 1985;100:686–697.
94. Green WR. Clinicopathologic studies of senile macular degeneration. In: Nicholson DH, ed. Ocular
pathology update. Chicago, IL: Year Book Medical Publishers; 1982:115–144.
95. Hoshino M, Mizuno K, Ichikawa H. Aging alterations of retina and choroid of Japanese: light
microscopic study of macular region of 176 eyes. Jpn J Ophthalmol. 1984;28:89–102.
96. Ring HG, Fugino T. Observation on the anatomy and pathology of the choroidal vasculature. Arch
Ophthalmol. 1967;78:431–444.
97. Hogan MJ. Role of the retinal pigment epithelium in macular disease. Trans Am Acad Ophthalmol
Otolaryngol. 1972;76(1):64–80.
4
ETIOLOGY OF LATE AGERELATED
MACULAR DISEASE
MAXIMILIANO OLIVERA
INTRODUCTION
Age-related macular disease (AMD) is a chronic, progressive disease of the
retina, specifically the macula. AMD is an important cause of vision loss in the
United States and all over the world. AMD, in all of its forms, has an overall
prevalence of 6.5%, but some demographics, such as non-Hispanic White
people, have a higher incidence (7.3%). Non-Hispanic Black people have a
lower incidence rate (2.4%). The incidence of AMD increases with age. The
incidence in individuals over 60 years old is 13.4% compared to an incidence in
the 40- to 59-year-old group of 2.8% (1).
Like many other chronic diseases, it is important to understand its complex
pathophysiology in order to develop screening and treatment strategies.
Although we do not have a complete understanding of the complex
pathophysiology of AMD, several studies over the last 20 years have identified
many environmental and genetic risk factors. Identification of risk factors allows
for the development of screening strategies in order to make a diagnosis as early
as possible, improving the chances of a better visual outcome. For example, the
understanding of pathophysiologic mechanisms involved in the development and
progression of neovascular AMD, such as the role of the vascular endothelial
growth factor (VEGF), permitted the design and development of anti-VEGF
therapy for AMD. This has resulted in a radical change in the expectations for
patients and physicians (2).
CLINICAL CLASSIFICATION OF AMD
AMD is not only about changes in the eye fundus examination. To diagnose
AMD and determine what kind of AMD the patient has, it is important to
consider the patient's symptoms.
In addition, it is important to understand that the aging eye is associated with
certain changes (3). Most of these changes are almost clinically undetectable,
and patients have no visual symptoms at all. These changes include the
following:
Early AMD
Early AMD is usually asymptomatic and clinically presents with drusen and
RPE changes consisting of focal hyper- or hypopigmentation.
Late AMD
The main characteristic of late AMD is severe loss of vision.
The late stages of AMD have two different forms (dry and wet), although
both forms may occur simultaneously in the same eye. Over time, dry AMD may
develop to wet AMD.
Dry AMD
Patients with dry AMD have poor visual acuity, hyperpigmented RPE spots
(usually juxtafoveal), and an area with visible choroidal vessels. Choroidal
vessels are visible because the RPE cells over them have become atrophic. As
these areas become confluent, they take on an appearance referred to as
geographic atrophy. No neovascularization is present.
Wet AMD
Wet AMD is characterized by the presence of CNVM. Patients with wet AMD
may experience a sudden onset of decreased visual acuity, metamorphopsia, and
paracentral scotoma. Clinical findings include subretinal fluid, subretinal or sub-
RPE hemorrhages, subretinal or intraretinal lipids, pigmentary subretinal ring,
RPE irregularities and detachment, gray-white subretinal lesions, and cystoid
macular edema (3).
EPIDEMIOLOGY
Incidence of AMD
According to the 2005–2008 NHANES (National Health and Nutrition
Examination Survey) (1), the most AMD-compatible lesions were found in
persons aged 60 or older in all the three ethnic groups studied (non-Hispanic
Black, non-Hispanic White, and Mexican American). The incidence of early
AMD is similar for non-Hispanic White persons aged 40 to 59 years (3%) and
Mexican American persons aged 40 to 59 years (2.7%). The incidence of early
AMD is lowest in non-Hispanic Black persons. Late AMD was more prevalent
in non-Hispanic White persons.
The estimated total prevalence of any AMD in the US population aged 40 or
older was 6.5%. Of a total of 7.2 million persons having any kind of AMD, 0.89
million (95% CI, 552,000–1.2 million) were estimated to have late AMD (see
Ref. (1) for more details).
GENETICS OF AMD
Patients with a positive familiar history of AMD have a higher risk of
development of this disease (8,25,26). Late AMD is a polygenic disease. No
single-gene defect accounts for disease. There are variations and defects in genes
functionally related to complement and immune processes, high-density
lipoprotein (HDL) cholesterol, and mechanisms involving collagen formation,
extracellular matrix production, and angiogenesis considered to be associated
with the onset, progression, and bilateral involvement of early, intermediate, and
advanced states of AMD (see Table 4.1 and Refs. (27–34) for more details) (35).
Several pharmacogenetic studies found that variants in some genes are related to
different treatment outcomes (Table 4.2) (36).
Adapted from Lim LS, Mitchell P, Seddon JM, et al. Age-related macular degeneration. Lancet.
2012;379:1728–1738.
Table 4.2 PHARMACOGENETICS AND AMD
Adapted from Chen Y, Bedell M, Zhang K. Age-related macular degeneration: genetic and environmental
factors of disease. Mol Interv. 2010;10(5):271–281.
Complement Factor H
Complement factor H (CFH), an important regulator of the alternative pathway,
was the first complement protein to be implicated in the pathogenesis of AMD.
Physiologically, CFH binds C3b and accelerates the decay of the alternative C3
convertase (C3bBb) and acts as cofactor for the inactivation of C3b by
complement factor I (CFI) (37). CFH is produced locally in the eye by the RPE
cells (38) and accumulates in the drusen, sub-RPE space, RPE,
interphotoreceptor matrix, and choroid (38,39). Environmental factors vary CFH
production by the RPE cells. In vitro studies show an increase in RPE CFH
production by interferon gamma (40) and reduction in conditions of oxidative
stress (38).
The CFH gene is located at chromosome position 1q23. Mutations of this
gene manifest as dominant mendelian disorders (41). In 2005, three groups
simultaneously (27,42,43) reported a nonsynonymous SNP in the CFH gene
(rs1061170), resulting in a substitution of tyrosine by histidine at position 402 of
the polypeptide (Y402H), which was important in the development of AMD.
This change alters the ligand binding site of CRP (C-reactive protein), heparin,
M protein, and glycosaminoglycans, probably leading to a reduced binding to
cell surfaces and therefore impaired regulation of the alternative C3 convertase
(44). A meta-analysis (45) suggested that this variant (Y402H) is a contributing
factor in over half of all cases of AMD. Other SNPs throughout the CFH gene,
including the SNP 162 V (38), resulting in substitution of isoleucine with valine
residue within the C3b binding site, have been associated with AMD
(32,38,41,46–50) and AMD progression (51–55).
The reconstitution of the visual pigment rhodopsin occurs mainly in the RPE
cells, through many intermediate steps. This mechanism has a key role for
normal function of both cones and rods, transforming the all-trans-retinyl esters
into 11-cis-retinal (65). As a phagocytic system, the RPE is essential for the
renewal of PRs (66). Each PR has hundreds of disks in its outer segment. These
disks are formed by plasma membrane, containing transmembrane protein
rhodopsin, which is positioned in combination with four phospholipids and
docosahexanoic acid. The OS disks of the PR cells are engulfed by the RPE. In
the RPE, the disks fuse with lysosomes, forming phagolysosomes, where the
contents are degraded. In young, healthy individuals, most of the disks are fully
degraded, and lipofuscin accumulation is minimal, but over time, the self-limited
phagocytic and degradative capacity of the RPE cells becomes more and more
overloaded. This incompletely degraded membrane material accumulates in the
form of lipofuscin in secondary lysosomes or residual bodies (67).
Lipofuscin is a yellow-brown, autofluorescent molecule that accumulates in
all postmitotic cells, particularly the RPE (68–71). The presence of lipofuscin
may act as a cellular aging indicator, and its quantity in tissues may be estimated
by amounts of autofluorescence present. The autofluorescence from the eye
fundus, mostly derived from lipofuscin, can be clinically and noninvasively
quantified, allowing for an estimate of the aging degenerative process of the eye
and a diagnostic and follow-up method for patients with AMD (72). Some
factors increase (vitamin E deficiency) while others decrease (oxygen-free
conditions and vitamin A deficiency) lipofuscin pigment formation.
The retinoid A2E is the major fluorophore of lipofuscin (73). Once it is
synthesized, it cannot be eliminated by the RPE. Precursors of A2E, all-trans-
retinal and ethanolamine, are formed within the PRs (74), but the fully
synthesized A2E molecule arises from the phagolysosomal compartment of the
RPE cells. When it reaches a critical concentration, the metabolism of
phagocytized OS lipids by the RPE is impaired. Phagocytized and oxidized OS
membranes are extruded by the RPE into the Bruch's membrane, contributing to
drusen formation and membrane thickening.
A2E is toxic for the RPE. It inhibits the proton pump of lysosomes (75),
causing leakage of the contents of the lysosomes into the cytoplasm of RPE
cells. It inhibits phagolysosomal degradation of PR phospholipids (76) and can
also damage the DNA of RPE cells. A2E also accumulates in the mitochondrial
membranes, decreasing mitochondrial activity and enabling the translocation of
cytochrome C and AIF (apoptosis-inducing factor) to the cytosol and nucleus,
respectively. Functionally, the release of cytochrome C from the inner
mitochondrial membrane generates oxidative stress and decreased electron flow,
leading to impaired ATP synthesis. Both mechanisms are highly relevant for
apoptosis by causing leakiness of the inner mitochondrial membrane and release
of the propapoptotic proteins, activating the caspase cascade. AIF is a pro-
apoptotic protein, which is strictly located in the mitochondria. Its translocation
to the nucleus induces apoptosis, functionally independent from caspases (77).
A2E also inhibits the normal activity of the enzyme RPE65 isomerohydrolase, a
key enzyme of the visual cycle, which is responsible for the isomerization of all-
trans-retinyl ester to 11-cis-retinol, the precursor of 11-cis-retinal (78). This
inhibition could cause a disruption of 11-cis-retinal supply to the retina and
result in PR dysfunction. A2E is known to perturb the cholesterol metabolism in
the RPE cells, causing cholesterol accumulation within the late
endosome/lysosome, inhibiting acid lipase and resulting in a feed-forward cycle
of cholesterol accumulation in the RPE. This interferes with rhodopsin activation
and PR membrane remodeling (79). A2E is also thought to be photosensitizing
due to its broad light spectrum–absorbing capacities, particularly in the visible
ranges, and thus generating reactive species of oxygen (80,81).
Choriocapillaris
The choriocapillaris is the vascular layer of the eye. Between the retina and the
sclera, it is formed by an extensive fenestrated vascular network derived from
the perforating ciliary arteries, branches of the ophthalmic artery. Its main role is
to provide oxygen and macromolecules to the outer retina. Over the years, the
choriocapillaris becomes less fenestrated and the lumina are reduced, causing
less delivery of oxygen and macromolecules to the retina (98,108,109). This
change results in hypoxia and activation in the transcription of genes such as
erythropoietin (for PR protection) (110) and vascular endothelial growth factor A
(VEGF-A) (111,112), causing the development of CNVM.
Angiogenesis
Angiogenesis, leading to the formation of CNVM, is one of the most important
pathophysiologic features of late AMD. In angiogenesis, blood vessels grow in
adult tissue from sprouts on preexisting vessels. This process involves several
steps: cell migration, proliferation, and survival; vascular maturation; wall
remodeling; and degradation of the extracellular matrix (114–116). Angiogenesis
has a complex molecular regulation, representing a balance between the
angiogenic and the antiangiogenic factors, affected by different pathologic states
such as hypoxia, ischemia, or inflammation (2).
Hypoxia
The role of hypoxia on the pathogenesis of AMD is controversial. While some
studies (123) showed that patients with AMD have decreased blood flow in the
choriocapillaris, others (124) could not confirm a decrease in blood flow but
found a decrease in pulse amplitude. It also has been theorized that the
previously described age-related changes in the Bruch's membrane may limit the
diffusion of oxygen and create an ischemic environment. Physiologically, pO2
decreases linearly with distance from the choriocapillaris to the inner segment of
the PRs. Under normal conditions, little of the O2 in the choriocapillaris blood is
extracted. Some studies have shown that as perfusion decreases, the oxygen
extraction from the choriocapillaris increases (125), showing that there is
significant reserve. This suggests that hypoxia is not the most important factor in
causing an imbalance between angiogenic and antiangiogenic factors.
Inflammation
Studies of excised choroidal membranes and the growth patterns of CNVM
suggest more is involved than just ischemia in driving neovascularization.
Histologic studies of the CNVM show vascular endothelium as well as a variety
of inflammatory cells, such as lymphocytes, macrophages, and foreign body
giant cells (126,127), similar to granulation tissue on a wound-repair response
(128). Activated macrophages secrete proteolytic enzymes, collagenase, and
elastase, which can erode an attenuated Bruch's membrane and thereby assist the
migration of choroidal capillaries (129,130). Also, inflammatory cells produce
cytokines such as interleukin-1, which induce VEGF, as shown in cultured
choroidal fibroblasts (131). In one study (132), the amount of VEGF was found
to be proportional to the number of macrophages in the specimen, suggesting
that inflammation is important for the regulation of angiogenesis. Therefore,
upregulation of VEGF may be the link between inflammation and promotion of
new blood vessels in an eye with AMD.
Before the formation of the CNVM, there are signs of inflammation in an
eye with AMD. Drusen deposits between the Bruch's membrane and RPE may
contain bioactive fragments of complements (C3a and C5a) (133). These
substances induce VEGF expression and have significant chemotactic activity
that further invites inflammatory cells to the macular region (134). Another key
actor of the complement system in AMD is the CFH. CFH binds and inactivates
C3b deposited on intact host cells, allowing destruction of foreign or damaged
cells (135). This is achieved through an important locus, domain 7 on the
molecule (136–139). Domain 7 binds to heparin or sialic acid on host cells.
Therefore, it is possible that alterations in domain 7 or the heparin/or sialic acid
binding region of factor H could lead to or augment the destruction of normal or
injured ocular cells as well as other foreign cells in an appropriate environment
(44).
Extracellular drusen also contain advanced glycation end products (AGE),
high levels of oxidized low-density lipoproteins (ox-LDL), and oxysterols
(87,140,141). The receptor for AGE (RAGE), normally not markedly expressed
in the retina, although highly accumulated in RPE cells, PRs, and
choriocapillaris in advanced AMD (142,143), activates the nuclear factor kappa
B (NFκB), master regulator of both innate and adaptive immunity. NFκB is a
central regulator of IL-6, a cytokine that has been shown to be an important
regulator of choroidal neovascularization (144–146), and VEGF. Damaged RPE
and oxidized proteins and lipids in the Bruch's membrane have been postulated
to activate and recruit dendritic cells from the choroid, enhancing the immune
response in this area (88,147).
In a laser model of CNV, CD18 and ICAM-1 are expressed during
development of CNV. Targeted disruption of either of these inhibits the
development of CNVM. Animals models of CNV have been developed that
mimic many aspects of CNVM in AMD. Mice expressing monocyte
chemoattractant protein-1 (MCP-1) or its cognate, CC chemokine receptor-2,
developed drusen, lipofuscin accumulation, geographic atrophy, and CNV (148).
Depletion of the monocyte cell lines inhibits experimental CNV (149–151).
With time, the neovascularization appears to “burn out,” leaving a cicatricial
mass almost completely devoid of vessels. If ischemia is the only cause of
neovascularization, one would expect these vessels to regress, which would
increase the amount of ischemia present.
The Complement System
The complement system plays a fundamental role in host defense against
pathogens, elimination of immune complexes and apoptotic cells, and adaptive
immune responses. It consists of over 40 proteins and regulators found in the
systemic circulation and some tissues. Three different activation pathways exist,
each one with different triggers: the classical pathway (triggered by an antibody–
antigen complex), the alternative pathway (triggered by binding to a host cell
pathogen surface), and the lectin pathway (triggered by polysaccharides on
microbial surfaces). These three pathways converge on the terminal complement
complex (TCC), a multiprotein complex, formed from the binding of C5b to the
plasma complement proteins C6, C7, C8, and C9 (152).
Components of the classic, alternative complement pathways and TCC have
been found to be present in drusen (153), especially in older eyes with AMD.
Alterations of the complement system or its regulators can lead to damage of
the tissues and is known to be implicated in a wide spectrum of diseases. Several
studies suggest that complement plays a key role in the development of AMD
(154).
Oxidative Stress
Oxidative stress refers to a progressive cellular damage caused by reactive
oxygen species (ROS) contributing to protein misfolding and evoking functional
abnormalities during RPE cellular senescence (172). Light can induce the
formation of reactive species, which in turn can lead to the formation of
dysfunctional or toxic molecules of lipids and proteins or damage the DNA.
Oxidized molecules are formed in the PR OSs as a normal part of daily life.
Over the years, the amount of these molecules increases because the mechanisms
needed to clear the reactive species out of the cells become insufficient (173).
Oxidative stress also interferes with the capacity of the heat shock proteins and
the ubiquitin–proteasome way to repair or degrade damaged proteins (174–176).
Aging primarily influences the oxidative balance between oxidizing factors
and antioxidants. Other factors, such as cigarette smoking and alcohol
consumption, aggravate oxidative imbalance and DNA damage (177).
Antioxidant genes (178) and variants of the CFH gene influence the role of
oxidative stress in AMD (179).
A wide variety of oxidative lesions to DNA have been described, including
single- and double-stranded DNA breaks and the development of cross-linked
lesions (180,181). The cell responds to genomic damage through repair
processes employing a large number of proteins. The cell may turn off growth
and replication until the repair process is complete (182). Responding to the
Samurai law of biology (better dead than wrong), those cells unable to repair
their DNA damage undergo apoptosis.
To help protect against inappropriate oxidation, there are basically three
levels of protection: molecular, cellular, and tissue. On the molecular level, the
cell has antioxidant vitamins and enzymes. These include vitamins C and E,
superoxide dismutase, catalase, glutathione transferase (183), glutathione
reductase, and glutathione peroxidase. The antioxidants may limit inappropriate
oxidation in the first place or may terminate propagation reactions. On a cellular
level, two main responses may occur. The cell may try to adapt to the oxidative
stress by increased activation of transcription factors and proteins (184) that
helps control gene expression of antioxidant enzymes. The cell could also start
the apoptosis mechanism. Tissue mechanisms for responding or protecting
against oxidative stress include complex intercellular signaling pathways as
discussed below.
Scavenger receptors (185,186), in particular CD36, are present on the RPE
cell and participate in phagocytosis of spent OSs. It is possible that ordinary
everyday exposure of CD36 receptor to oxidized lipids in the PR OS helps
maintain the constitutive secretion of VEGF by RPE cells. Excessive secretion
of VEGF by RPE cells is supposed to be one factor responsible for the initiation
of CNV. This raises the possibility that excessive exposure to oxidative damage
may lead the RPE cells to secrete excessive VEGF. Armstrong et al. (187) have
shown that injection of lipid peroxide derivate into the vitreous cavity caused
retinal neovascularization that persisted for 4 weeks. After the injection, there
was a cascade of cytokines secreted, including VEGF. The Bruch's membrane
has an exponential increase in lipids with age. The lipids preferentially
accumulate in the same region where the neovascularization grows. The Bruch's
membrane has no known intrinsic mechanism to protect against oxidative
damage to lipids, which accumulate over time. This mechanism could be
important in the formation of CNVM in AMD.
Lipid peroxides not only accumulate in the Bruch's membranes but also
accumulate in atherosclerotic plaques. In atherosclerotic vessels, the body
mounts an aggressive cell-mediated mechanism to contain the oxidized material
(188,189), principally using vascular endothelial cells and macrophages also
stimulating the production of VEGF by these cells. The body has a number of
defined strategies and methods for dealing with degenerating cells and tissue.
Many of the same strategies and methods that are used in atherosclerosis of
vessel walls are also used in the eye. Injection of these lipids has led to ocular
neovascularization in the rabbit (190–192).
As discussed, the accumulation of oxidative damage may be a cause of
neovascularization, but the same oxidative damage may also induce a senescent
aging phenotype (193), with possible apoptosis. Oxidative damage can lead to an
increased accumulation of lipofuscin within RPE cells, a finding linked to the
development of geographic atrophy.
CONCLUSION
In this chapter, we discussed the etiology of age-related macular disease.
Epidemiologic and genetic factors, clinical classification, anatomical changes,
and how these changes affected the development and evolution of the AMD
were discussed. While a complete understanding of AMD and its
pathophysiology is still lacking, new evidence and understanding are rapidly
accumulating. There is not a single gene defect or a single molecular disturbance
that causes AMD. It is a complex, multifactorial disease that may require a
variety of treatment strategies to preserve vision for our patients.
REFERENCES
1. Klein R, Chou CF, Klein B, et al. Prevalence of age-related macular degeneration in the US Population.
Arch Ophthalmol. 2011;129(1):75–80.
2. Bressler SB. Introduction: understanding the role of angiogenesis and antiangiogenic agents in age-
related macular degeneration. Ophthalmology. 2009;116:S1–S7.
3. American Academy of Ophthalmology. Retina and Vitreous. Basic and Clinical Science Course Section
12. 2011–2012.
4. Age-Related Eye Disease Study Research Group. Risk factors for the incidence of advanced age-related
macular degeneration in the Age-Related Eye Disease Study (AREDS) AREDS Report No. 19.
Ophthalmology. 2005;112(4):533–539.
5. Klein R. Epidemiology. In: Berger JW, Fine SL, Maguire MG, eds. Age-related macular degeneration.
St. Louis, MO: Mosby; 1999:31–55.
6. Seddon J. Epidemiology of age-related macular degeneration. In: Schachat AR, ed. Medical retina, vol
2, 3rd Ed. St. Louis, MO: Mosby; 2001:1039–1050.
7. Smith W, Assink J, Klein R, et al. Risk factors for age-related macular degeneration: pooled findings
from three continents. Ophthalmology. 2001;108:697–704.
8. Hyman LG, Lilienfeld AM, Ferris FL III, et al. Senile macular degeneration: a case-control study. Am J
Epidemiol. 1983;118:213–227.
9. Christen WG, Glynn RJ, Ajani UA, et al. Age-related maculopathy in a randomized trial of low-dose
aspirin among US physicians. Arch Ophthalmol. 2001;119:1143–1149.
10. Klein R, Klein BE, Moss SE. Diabetes, hyperglycemia, and age-related maculopathy. The Beaver Dam
Eye Study. Ophthalmology. 1992;82:1527–1534.
11. Smith W, Mitchell P, Wang JJ. Gender, oestrogen, hormone replacement and age-related macular
degeneration: results from the Blue Mountains Eye Study. Aust N Z J Ophthalmol.
1997;25(Suppl):S13–S15.
12. Smith W, Mitchell P, Leeder SR, et al. Plasma fibrinogen levels, other cardiovascular risk factors, and
age-related maculopathy: the Blue Mountains Eye Study. Arch Ophthalmol. 1998;116:583–587.
13. Tomany SC, Wang JJ, van Leeuwen R, et al. Risk factors for incident age-related macular degeneration.
Pooled findings from 3 continents. Ophthalmology. 2004;111:1280–1287.
14. Snow KK, Cote J, Yang W, et al. Association between reproductive and hormonal factors and age-
related maculopathy in postmenopausal women. Am J Ophthalmol. 2002;134:842–848.
15. Edwards DR, Gallins P, et al. Inverse association of female hormone replacement therapy with age-
related macular degeneration and interactions with ARMS2 polymorphisms. Invest Ophthalmol Vis Sci.
2010;51(4):1873–1879.
16. Chumbley LC. Impressions of eye diseases among Rhodesian Blacks in Mashonaland. S Afr Med J
1977;52:316–318.
17. Gregor Z, Joffe L. Senile macular changes in the black African. Br J Ophthalmol. 1978;62:547–550.
18. Cho E, Hung S, Willet WC, et al. Prospective study of dietary fat and the risk of age-related macular
degeneration. Am J Clin Nutr. 2001;73:209–218.
19. Parekh N, Voland RP, et al; CAREDS Research Study Group. Association between dietary fat intake
and age-related macular degeneration in the Carotenoids in Age-Related Eye Disease Study (CAREDS): an
ancillary study of the Women's Health Initiative. Arch Ophthalmol. 2009;127(11):1483–1493.
20. Seddon JM, Rosner B, Sperduto RD, et al. Dietary fat and the risk for advanced age-related macular
degeneration. Arch Ophthalmol. 2001;119:1191–1199.
21. Seddon JM, Cote J, Rosner B. Progression of age-related macular degeneration: association with
dietary fat, transunsaturated fat, nuts and fish intake. Arch Ophthalmol. 2003;121:1728–1737.
22. Smith W, Mitchell P, Leeder SR. Dietary fat and fish intake and age-related maculopathy. Arch
Ophthalmol. 2000;118:401–404.
23. Seddon JM, Ajani UA, Sperduto RD, et al. Dietary carotenoids, vitamins A, C, and E, and advanced
age-related macular degeneration. Eye Disease Case Control Study Group. JAMA. 1994;272:1413–1420.
24. Guymer RH, Chong EW. Modifiable risk factors for age-related macular degeneration. Med J Aust.
2006;184(9):455–458.
25. Klaver CC, Wolfs RC, Assink JJ, et al. Genetic risk of age-related maculopathy. Population-based
familial aggregation study. Arch Ophthalmol. 1998;116;1646–1651.
26. Smith W, Mitchell P. Family history and age-related maculopathy: the Blue Mountains Eye Study. Aust
N Z J Ophthalmol. 1998;26:203–206.
27. Klein RJ, Zeiss C, Chew EY, et al. Complement factor H polymorphism in age-related macular
degeneration. Science. 2005;308:385–389.
28. Maller JB, Fagerness JA, Reynolds RC, et al. Variation in complement factor 3 is associated with risk
of age-related macular degeneration. Nat Genet. 2007;39:1200–1201.
29. Fagerness JA, Maller JB, Neale BM, et al. Variation near complement factor I is associated with risk of
advanced AMD. Eur J Hum Genet. 2009;17:100–104.
30. Reynolds R, Rosner B, Seddon JM. Serum lipid biomarkers and hepatic lipase gene associations with
age-related macular degeneration. Ophthalmology. 2010;117:1989–1995.
31. Neale BM, Fagerness J, Reynolds R, et al. Genome-wide association study of advanced age-related
macular degeneration identifies a role of the hepatic lipase gene (LIPC). Proc Natl Acad Sci U S A.
2010;107:7395–7400.
32. Chen W, Stambolian D, Edwards AO, et al. Genetic variants near TIMP3 and high-density lipoprotein-
associated loci influence susceptibility to age-related macular degeneration. Proc Natl Acad Sci U S A.
2010;107:7401–7406.
33. McKay GJ, Patterson CC, Chakravarthy U, et al. Evidence of association of APOE with age-related
macular degeneration—a pooled analysis of 15 studies. Hum Mutat. 2011;32:1407–1416.
34. Yu Y, Bhangale TR, Fagerness J, et al. Common variants near FRK/COL10A1 and VEGFA are
associated with advanced age-related macular degeneration. Hum Mol Genet. 2011;20:3699–3709.
35. Lim LS, Mitchell P, Seddon JM, et al. Age-related macular degeneration. Lancet.
2012;379:1728–1738.
36. Chen Y, Bedell M, Zhang K. Age-related macular degeneration: genetic and environmental factors of
disease. Mol Interv. 2010;10(5):271–281.
37. Pangburn MK. Host recognition and target differentiation by factor H, a regulator of the alternative
pathway of complement. Immunopharmacology. 2000;49:149–157.
38. Hageman GS, Anderson DH, Johnson LV, et al. A common haplotype in the complement regulatory
gene factor H (HF1/CFH) predisposes individuals to age-related macular degeneration. Proc Natl Acad Sci
U S A. 2005;102:7227–7232.
39. Chen M, Forrester JV, Xu H. Synthesis of complement factor H by retinal pigment epithelial cells is
down-regulated by oxidized photoreceptor outer segments. Exp Eye Res. 2007;84:635–645.
40. Kim YH, He S, Kase S, et al. Regulated secretion of complement factor H by RPE and its role in RPE
migration. Graefes Arch Clin Exp Ophthalmol. 2009;247:651–659.
41. Klein ML, Schultz DW, Edwards A, et al. Age-related macular degeneration. Clinical features in a
large family and linkage to chromosome 1q. Arch Ophthalmol. 1998;116:1082–1088.
42. Edwards AO, Ritter R III, Abel KJ, et al. Complement factor H polymorphism and age-related macular
degeneration. Science. 2005;308:421–424.
43. Haines JL, Hauser MA, Schmidt S, et al. Complement factor H variant increases the risk of age-related
macular degeneration. Science. 2005;308:419–421.
44. Khandhadia S, Cipriani V, Yates JRW, et al. Age-related macular degeneration and the complement
system. Immunobiology. 2012;217:127–146.
45. Thakkinstian A, Han P, McEvoy M, et al. Systematic review and meta-analysis of the association
between complement factor H Y402H polymorphisms and age-related macular degeneration. Hum Mol
Genet. 2006;15:2784–2790.
46. Li M, Atmaca-Sonmez P, Othman M, et al. CFH haplotypes without the Y402H coding variant show
strong association with susceptibility to age-related macular degeneration. Nat Genet. 2006;38:1049–1054.
47. Francis PJ, Schultz DW, Hamon S, et al. Haplotypes in the complement factor H (CFH) gene:
associations with drusen and advanced age-related macular degeneration. PLoS One. 2007;2:e1197.
48. Seddon JM, Reynolds R, Maller J, et al. Prediction model for prevalence and incidence of advanced
age-related macular degeneration based on genetic, demographic, and environmental variables. Invest
Ophthalmol Vis Sci. 2009;50:2044–2053.
49. Gibson J, Cree A, Collins A, et al. Determination of a gene and environment risk model for age-related
macular degeneration. Br J Ophthalmol. 2010;94:1382–1387.
50. Kopplin LJ, Igo RP Jr, Wang Y, et al. Genome-wide association identifies SKIV2L and MYRIP as
protective factors for age-related macular degeneration. Genes Immun. 2010;11:609–621.
51. Despriet DD, Klaver CC, Witteman JC, et al. Complement factor H polymorphism, complement
activators, and risk of age-related macular degeneration. JAMA. 2006;296:301–309.
52. Boekhoorn SS, Vingerling JR, Hofman A, et al. Alcohol consumption and risk of aging macular
disorder in a general population: the Rotterdam Study. Arch Ophthalmol. 2008;126:834–839.
53. Schaumberg DA, Christen WG, Kozlowski P, et al. A prospective assessment of the Y402H variant in
complement factor H, genetic variants in C-reactive protein, and risk of age-related macular degeneration.
Invest Ophthalmol Vis Sci. 2006;47:2336–2340.
54. Seddon JM, Francis PJ, George S, et al. Association of CFH Y402H and LOC387715 A69S with
progression of age-related macular degeneration. JAMA. 2007;297:1793–1800.
55. Farwick A, Wellmann J, Stoll M, et al. Susceptibility genes and progression in age-related
maculopathy: a study of single eyes. Invest Ophthalmol Vis Sci. 2010;51:731–736.
56. Haines JL, Schnetz-Boutaud N, Schmidt S, et al. Functional candidate genes in age-related macular
degeneration: significant association with VEGF, VLDLR, and LRP6. Invest Ophthalmol Vis Sci.
2006;47:329–335.
57. Churchill AJ, Carter JG, Lovell HC, et al. VEGF polymorphisms are associated with neovascular age-
related macular degeneration. Hum Mol Genet. 2006;15:2955–2961.
58. Lin JM, Wan L, Tsai YY, et al. Vascular endothelial growth factor gene polymorphisms in age-related
macular degeneration. Am J Ophthalmol. 2008;145:1045–1051.
59. Boltz A, Ruiß M, Jonas JB, et al. Role of vascular endothelial growth factor polymorphisms in the
treatment success in patients with wet age-related macular degeneration. Ophthalmology.
2012;119:1615–1620.
60. Taylor HR, West S, Munoz B, et al. The long-term effects of visible light on the eye. Arch Ophthalmol.
1992;110:99–104.
61. Kagan VE, Shvedova AA, Novikov KN, et al. Light-induced free radical oxidation of membrane lipids
in photoreceptors of frog retina. Biochim Biophys Acta. 1973;330:76–79.
62. Organisciak DT, Vaughan DK. Retinal light damage: mechanisms and protection. Prog Retin Eye Res.
2010;29:113–134.
63. Rodriguez-Rocha H, Garcia-Garcia A, Panayiotidis MI, et al. DNA damage and autophagy. Mutat Res.
2011;711:158–166.
64. De Jong PTVM. Age-related macular degeneration. N Engl J Med. 2006;355:1474–1485.
65. Redmond TM, Poliakov E, Yu S, et al. Mutation of key residues of RPE65 abolishes its enzymatic role
as isomerohydrolase in the visual cycle. Proc Natl Acad Sci U S A. 2005;102:13658–13663.
66. Young RW, Bok D. Participation of the retinal pigmentary epithelium in the rod outer segment renewal
process. J Cell Biol. 1969;42:392–403.
67. Eldred GE, Lasky MR. Retinal age pigments generated by self-assembling lysosomotropic detergents.
Nature. 1993;25(361):724–726.
68. Wolf G. Lipofuscin, the age pigment. Nutr Rev. 1993;51:205–206.
69. Boulton M, Marshall J. Effects of increasing number of phagocytic inclusions on human retinal
pigment epithelial cells in culture: a model for aging. Br J Ophthalmol. 1986;70:808–815.
70. FeeneyBurns L, Berman ER, Rothman H. Lipofuscin of human retinal pigment epithelium. Am J
Ophthalmol. 1980;90:783–791.
71. Wing GL, Blanchard GC, Weiter JJ. The topography and age relationship of lipofuscin concentration in
the retinal pigment epithelium. Invest Ophthalmol Vis Sci. 1978;17:601–607.
72. Delori FC, Dorey CK, Staurenghi G, et al. In vivo fluorescence of the ocular fundus exhibits retinal
pigment epithelium lipofuscin characteristics. Invest Ophthalmol Vis Sci. 1995;36:718–729.
73. Eldred GE. Lipofuscin fluorophore inhibits lysosomal protein degradation and may cause early stages
of macular degeneration. Gerontology. 1995;42:(Suppl 2):15–28.
74. Mata NL, Weng J, Travis GH. Biosynthesis of a major lipofuscin fluorophore in mice and humans with
ABCR-mediated retinal and macular degeneration. Proc Natl Acad Sci U S A. 2000;97:7154–7159.
75. Bergmann M, Schutt F, Holz FG, et al. Inhibition of the ATP-driven proton pump in RPE lysosomes by
the major lipofuscin fluorophore A2-E may contribute to the pathogenesis of age-related macular
degeneration. FASEB J. 2004;18:562–564.
76. Finnemann SC, Leung LW, Rodriguez-Boulan E. The lipofuscin component A2E selectively inhibits
phagolysosomal degradation of photoreceptor. Proc Natl Acad Sci U S A. 2002;99:6:3842–3847.
77. Suter M, Reme C, Grimm C, et al. Age-related macular degeneration: the lipofuscin component N-
retinyl-N-retinylidene ethanolamine detaches proapoptotic proteins from mitochondria and induces
apoptosis in mammalian retinal pigment epithelial cells. J Biol Chem. 2000;275:39625–39630.
78. Moiseyev G, Nikolaeva O, Chen Y, et al. Inhibition of the visual cycle by A2E through direct
interaction with RPE63 and implications in Stargardt disease. Proc Natl Acad Sci U S A.
2010;107:41:17551–17556.
79. Lakkaraju A, Finnemann S, Rodriguez-Boulan E. The lipofuscin fluorophore A2E perturbs cholesterol
metabolism in retinal pigment epithelial cells. Proc Natl Acad Sci U S A. 2007;104:26:11026–11031.
80. Sparrow JR, Zhou J, Cai B. DNA is a target of the photodynamic effects elicited in A2E-laden RPE by
blue-light illumination. Invest Ophthalmol Vis Sci. 2003;44:2245–2251.
81. Sparrow JR, Fishkin N, Zhou J, et al. A2E, a byproduct of the visual cycle. Vision Res.
2003;43:2983–2990.
82. Hewitt AT, Nakazawa K, Newsome DA. Analysis of newly synthesized Bruch's membrane
proteoglycans. Invest Ophthalmol Vis Sci. 1989;30:478–486
83. Kliffen M, de Jong PT, Luider TM. Protein analysis of human maculae in relation to age-related
maculopathy. Lab Invest. 1995;73:267–272.
84. Hogan MJ, Alvarado J. Studies on the human macula. IV. Aging changes in Bruch's membrane. Arch
Ophthalmol. 1967;77:410–420.
85. van der Schaft TL, de Bruijn WC, Mooy CM, et al. Is basal laminar deposit unique for age-related
macular degeneration? Arch Ophthalmol. 1991;109:420–425.
86. van der Schaft TL, Mooy CM, de Bruijn WC, et al. Histologic features of the early stages of age-related
macular degeneration: a statistical analysis. Ophthalmology. 1992;99:278–286.
87. Crabb JW, Miyagi M, Gu X, et al. Drusen proteome analysis: an approach to the etiology of age-related
macular degeneration. Proc Natl Acad Sci U S A. 2002;99:14682–14687.
88. Hageman GS, Luthert PJ, Victor Chong NH, et al. 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 Retin Eye Res. 2001;20:705–732.
89. Rudnew A. Ueber die Entstehung, der sogenannten Glaskoerper der Choroides des menschlichen
Auges und über das Wesen der hyalinen Degeneration der Gefässe derselben. Arch Pathol Anat Phyiol Klin
Med. 1871;53:455–465.
90. Nakata K, Crabb JW, Hollyfield JG. Crystallin distribution in Bruch's membrane-choroid complex
from age-related macular degeneration and age-matched donor eyes. Exp Eye Res. 2005;80:821–826.
91. Umeda S, Suzuki MT, Okamoto H, et al. Molecular composition of drusen and possible involvement of
anti-retinal autoimmunity in two different forms of macular degeneration in cynomolgus monkey (Macaca
fascicularis). FASEB J. 2005;19:2088.
92. Green WR, Enger C. Age-related macular degeneration histopathologic studies. The 1992 Lorenz E.
Zimmerman Lecture. Ophthalmology. 1993;100:1519–1535.
93. Green WR. Histopathology of the age related macular degeneration. Mol Vis. 1999;3;5–27.
94. Grossniklaus HE, Green WR. Histopathologic and ultrastructural findings of surgically excised
choroidal neovascularization. Submacular Surgery Trials Research Group. Arch Ophthalmol.
1998;116:745–749.
95. Ishibashi T, Sorgente N, Patterson R, et al. Aging changes in Bruch's membrane of monkeys: an
electron microscopy study. Ophthalmologica. 1986;192:179–190.
96. Van der Schaft TL, Mooy CM, de Brujin WC, et al. Immunohistochemical light and electron
microscopy of basal laminar deposit. Graefes Arch Clin Exp Ophthalmol. 1994;232:40–46.
97. Sarks JP, Sarks SH, Killingsworth MC. Evolution of soft drusen in age-related macular degeneration.
Eye. 1994;8(Pt 3):269–283.
98. Ramrattan RS, van der Schaft TL, Mooy CM, et al. Morphometric analysis of Bruch's membrane, the
choriocapillaris, and the choroid in aging. Invest Ophthalmol Vis Sci. 1994;35:2857–2864.
99. Bird AC, Marshall J. Retinal pigment epithelial detachments in the elderly. Trans Ophthalmol Soc U K.
1986;105:674–682.
100. Pauleikhoff D, Harper CA, Marshall J, et al. Aging changes in Bruch's membrane: a histochemical
and morphologic study. Ophthalmology. 1990;97: 171–178.
101. Holz FG, Sheraidah G, Pauleikhoff D, et al. Analysis of lipid deposits extracted from human macular
and peripheral Bruch's membrane. Arch Ophthalmol. 1994;112:402–406.
102. Marshall J, Hussain AA, Starita C, et al. Aging and Bruch's membrane. In: Marmor MF,
Wolfensberger TJ, eds. The retinal pigment epithelium: function and disease. New York: Oxford University
Press; 1998:669–692.
103. Lyda W, Eriksen N, Krishna N. Studies of Bruch's membrane: flow and permeability studies in a
Bruch's membrane-choroid preparation. Am J Ophthalmol. 1957;44:362–369.
104. Starita C, Hussain AA, Pagliarini S, et al. Hydrodynamics of ageing Bruch's membrane: implications
for macular disease. Exp Eye Res. 1996;62:565–572.
105. Curcio CA, Millican CL, Bailey T, et al. Accumulation of cholesterol with age in human Bruch´s
membrane. Invest Ophthalmol Vis Sci. 2001;42:265–274.
106. Gilmore AP. Anoikis. Cell Death Differ. 2005;12:(Suppl 2):1473–1477.
107. Bertrand K. Survival of exfoliated epithelial cells: a delicate balance between anoikis and apoptosis. J
Biomed Biotech. 2011;2011:1–9.
108. Sarks SH. Changes in the region of the choriocapillaris in ageing and degeneration. In: Shimizu K, ed.
XXIII Concilium Ophthalmologicum, Kyoto 1978, Acta, vol 1. Amsterdam, The Netherlands: Excerpta
Medica; 1979:228–238.
109. Korte GE, Reppucci V, Henkind P. RPE destruction causes choriocapillary atrophy. Invest
Ophthalmol Vis Sci. 1984;25:1135–1145.
110. Grimm C, Wenzel A, Groszer M, et al. HIF-1-induced erythropoietin in the hypoxic retina protects
against light-induced retinal degeneration. Nat Med. 2002;8:718–724.
111. D'Amore PA. Mechanisms of retinal and choroidal neovascularization. Invest Ophthalmol Vis Sci.
1994;35:3974–3979.
112. Spilsbury K, Garrett KL, Shen WY, et al. Overexpression of vascular endothelial growth factor
(VEGF) in the retinal pigment epithelium leads to the development of choroidal neovascularization. Am J
Pathol. 2000;157:1413.
113. Feund KB, Zweifel SA, Engelbert M. Do we need a new classification for choroidal
neovascularization in age-related macular degeneration?. Retina. 2010;30:9:1333–1349.
114. Ferrara N, Kerbel RS. Angiogenesis as a therapeutic target. Nature. 2005;438:967–974.
115. Pepper MS. Manipulating angiogenesis. From basic science to the bedside. Arterioscler Thromb Vasc
Biol. 1997;17:605–619.
116. Bernanke DH, Velkey JM. Development of the coronary blood supply: changing concepts and current
ideas. Anat Rec. 2002;269:198–208.
117. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003;9:669–676.
118. Wang F, Rendahl KG, Manning WC, et al. AAV-mediated expression of vascular endothelial growth
factor induces choroidal neovascularization in rat. Invest Ophthalmol Vis Sci. 2003;44:781–790.
119. Terman BI, Dougher-Vermazen M, Carrion ME, et al. Identification of the KDR tyrosine kinase as a
receptor for vascular endothelial cell growth factor. Biochem Biophys Res Commun. 1992;187:1579–1586.
120. Blaauwgeers HGT, Holtkamp GM, Rutten H. et al. Polarized vascular endothelial growth factor
secretion by human retinal pigment epithelium and localization of vascular endothelial growth factor
receptors on the inner choriocapillaris: evidence for a trophic paracrine relation. Am J Pathol.
1999;155:421–428.
121. Shima DT, Nishijima K, Jo N, et al. VEGF-mediated neuroprotection in ischemic retina. Invest
Ophthalmol Vis Sci. 2004;45:3270.
122. Kim I, Ryan A, Rohan R, et al. Constitutive expression of VEGF, VEGFR-1, and VEGFR-2 in normal
eyes. Invest Ophthalmol Vis Sci. 1999;40:2115–2121.
123. Grunwald JE, Hariprasad SM, DuPont J, et al. Foveolar choroidal blood flow in age-related macular
degeneration. Invest Ophthalmol Vis Sci. 1998;39:385–390.
124. Mori F, Konno S, Hikichi T, et al. Pulsatile ocular blood flow study; decreases in exudative age
related macular degeneration. Br J Ophthalmol. 2001;85:531–533.
125. Lisenmeier RA, Braun RD. Oxygen distribution and consumption in the cat retina during normoxia
and hypoxemia. J Gen Physiol. 1992;99:177–197.
126. Penfold PI, Killingsworth MC, Sarks SH. Senile macular degeneration: the involvement of
immunocompetent cells. Graefes Arch Clin Exp Ophthalmol. 1985;223:69–76.
127. Penfold PI, Killingsworth MC, Sarks SH. Senile macular degeneration. The involvement of giant cells
in atrophy of the retinal pigment epithelium. Invest Ophthalmol Vis Sci. 1986;27:364–371.
128. Spraul CW, Lang GE, Grossniklaus HE, et al. Histologic and morphometric analysis of the choroid,
Bruch´s membrane, and retinal pigment epithelium in postmortem eyes with age-related macular
degeneration and histologic examination of surgically excised choroidal neovascular membranes. Surv
Ophthalmol. 1999;44(Suppl):S10–S32.
129. Werb Z, Gordon S. Secretion of a specific collagenase by stimulated macrophages. J Exp Med.
1975;142:346–360.
130. Unanue ER, Beller DI, Calderon J, et al. Regulation of immunity and inflammation by mediators from
macrophages. Am J Pathol. 1976;85:465–478.
131. Kvanta A, Algvere PV, Berglin L, et al. Subfoveal fibrovascular membranes in age-related macular
degeneration express vascular endothelial growth factor. Invest Ophthalmol Vis Sci. 1996;37:1929–1934.
132. Ku HH, Sohal RS. Comparison of mitochondrial pro-oxidant generation and antioxidant defenses
between rat and pigeon: possible basis of variation in longevity and metabolic potential. Mech Ageing Dev.
1993;72:67–76.
133. Nozaki M, Raisler BJ, Sakurai E, et al. Drusen complement components C3a and C5a promote
choroidal neovascularization. Proc Natl Acad Sci U S A. 2006;103:2328–2333.
134. Kijlstra A, La HE, Hendrikse F. Immunological factors in the pathogenesis and treatment of age-
related macular degeneration. Ocul Immunol Inflamm. 2005;13:3–11.
135. Donoso LA, Kim D, Frost A, et al. The role of inflammation in the pathogenesis of age-related
macular degeneration. Surv Ophthalmol. 2006;51:137–152.
136. Giannakis E, Male DA, Ormsby RJ, et al. Multiple ligand binding sites on domain seven of human
complement factor H. Int Immunopharmacol. 2001;1:433–443.
137. Goldberg AF, Molday RS. Defective subunit assembly underlies a digenic form of retinitis
pigmentosa linked to mutations in peripherin/rds and rom-1. Proc Natl Acad Sci U S A.
1996;93:13726–13730.
138. Goodrich DW, Lee WH. The molecular genetics of retinoblastoma. Cancer Surv. 1990;9:529–554.
139. Gordon DL, Kaufman RM, Blackmore TK, et al. Identification of complement regulatory domains in
human factor H. J Immunol. 1995;155:348–356.
140. Ishibashi T, Murata T, Hangai M, et al. Advanced glycation end products in age-related macular
degeneration. Arch Ophthalmol. 1998;116:1629–1632.
141. Javitt NB, Javitt JC. The retinal oxysterol pathway: a unifying hypothesis for the cause of age-related
macular degeneration. Curr Opin Ophthalmol. 2009;20:151–157.
142. Yamada Y, Ishibashi K, Ishibashi K, et al. The expression of advanced glycation end-product
receptors in rpe cells associated with basal deposits in human maculas. Exp Eye Res. 2006;82:840–848.
143. Howes KA, Liu Y, Dunaief JL, et al. Receptor for advanced glycation endproducts and age-related
macular degeneration. Invest Ophthalmol Vis Sci. 2004;45:3713–3720.
144. Izumi-Nagai K, Nagai N, Ozawa Y, et al. Interleukin-6 receptor-mediated activation of signal
transducer and activator of transcription-3 (STAT3) promotes choroidal neovascularization. Am J Pathol.
2007;170:2149–2158.
145. Koto T, Nagai N, Mochimaru H, et al. Eicosapentaenoic acid is anti-inflammatory in preventing
choroidal neovascularization in mice. Invest Ophthalmol Vis Sci. 2007;48:4328–4334.
146. Paimela T, Ryhanen T, Mannermaa E, et al. The effect of 17b-estradiol on IL-6 secretion and NF-jB
DNA-binding activity in human retinal pigment epithelial cells. Immunol Lett. 2007;110:139–144.
147. Johnson LV, Leitner WP, Staples MK, et al. Complement activation and inflammatory processes in
drusen formation and age related macular degeneration. Exp Eye Res. 2001;73:887–896.
148. Ambati J, Anand A, Fernandez S, et al. An animal model of age-related macular degeneration in
senescent Ccl2- or Ccr2deficient mice. Nat Med. 2003;9:1390–1397.
149. Espinosa-Heidmann DG, Suner IJ, Hernandez EP, et al. Macrophage depletion diminishes lesion size
and severity in experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2003;44:3586–3592.
150. Sakurai E, Anand A, Ambati BK, et al. Macrophage depletion inhibits experimental choroidal
neovascularization. Invest Ophthalmol Vis Sci. 2003;44:3578–3585.
151. Ishida S, Usui T, Yamashiro K, et al. VEGF164-mediated inflammation is required for pathological,
but not physiological, ischemia induced retinal neovascularization. J Exp Med. 2003;198:483–489.
152. Comp Zhou W, Marsh JE, Sacks SH. Intrarenal synthesis of complement. 2011.
http://www.nature.com/ki/journal/v59/n4/fig tab/4492147f1.html.
153. Mullins RF, Russell SR, Anderson DH, et al. Drusen associated with aging and age-related macular
degeneration contain proteins common to extracellular deposits associated with atherosclerosis, elastosis,
amyloidosis, and dense deposit disease. FASEB J. 2000;14:835–846.
154. Anderson DH, Mullins RF, Hageman GS, et al. A role for local inflammation in the formation of
drusen in the aging eye. Am J Ophthalmol. 2002;134:411–431.
155. Dawson DW, Volpert OV, Gillis P, et al. Pigment epithelium-derived factor: a potent inhibitor of
angiogenesis. Science. 1999;285:245–248.
156. Spranger J, Osterhoff M, Reimann M, et al. Loss of the antiangiogenic pigment epithelium-derived
factor in patients with angiogenic eye disease. Diabetes. 2001;50:2641–2645.
157. Holekamp NM, Bouck N, Volpert O. Pigment epithelium-derived factor is deficient in the vitreous of
patients with choroidal neovascularization due to age-related macular degeneration. Am J Ophthalmol.
2002;134:220–227.
158. Ek ET, Dass CR, Choong PF. Pigment epithelium-derived factor: a multimodal tumor inhibitor. Mol
Cancer Ther. 2006;5:1641–1646.
159. Campochiaro PA, Glaser BM. Platelet-derived growth factor is chemotactic for human retinal pigment
epithelial cells. Arch Ophthalmol. 1985;103:576–579.
160. Choudary P, Chen W, Hunt R. Production of platelet-derived growth factors by interleukin-1b and
transforming growth factor-b stimulate RPE cells and leads to contraction of collagen gels. Invest
Ophthalmol Vis Sci. 1997;38:824–833.
161. De Oliveira Dias JR, Buchele Rodrigues E, Maia M, et al. Cytokines in neovascular age-related
macular degeneration: fundamentals of targeted combination therapy. Br J Ophthalmol.
2011;95:1631–1637.
162. Martin TA, Mansel R, Jiang WG. Hepatocyte growth factor modulates vascular endothelial-cadherin
expression in human endothelial cells. Clin Cancer Res. 2001;7:734–737.
163. Rosenthal R, Malek G, Salomon N, et al. The fibroblast growth factor receptors, FGFR-1 and FGFR-
2, mediate two independent signalling pathways in human retinal pigment epithelial cells. Biochem Biophys
Res Commun. 2005;337:241–247.
164. García M, Vecino E. Role of Müller glía in neuroprotection and regeneration in the retina. Histol
Histopathol. 2003;18:1205–1218.
165. Hollborn M, Jahn K, Limb GA, et al. Characterization of the basic fibroblast growth factor-evoked
proliferation of the human Müller cell line, MIO-M1. Graefes Arch Clin Exp Ophthalmol.
2004;242:414–422.
166. Stahl A, Paschek L, Martin G, et al. Combinatory inhibition of VEGF and FGF2 is superior to solitary
VEGF inhibition in an in vitro model of RPE-induced angiogenesis. Graefes Arch Clin Exp Ophthalmol.
2009;247:767–773.
167. Hera R, Keramidas M, Peoc'h M, et al. Expression of VEGF and angiopoietins in subfoveal
membranes from patients with age-related macular degeneration. Am J Ophthalmol. 2005;139:589–596.
168. Pichiule P, Chavez JC, LaManna JC. Hypoxic regulation of angiopoietin-2 expression in endothelial
cells. J Biol Chem. 2004;279:12171–12180.
169. Maisonpierre PC, Suri C, Jones PF, et al. Angiopoietin-2, a natural antagonist for Tie2 that disrupts in
vivo angiogenesis. Science. 1997;277:55–60.
170. Hoffmann S, He S, Ehren M, et al. MMP-2 and MMP-9 secretion by RPE is stimulated by angiogenic
molecules found in choroidal neovascular membranes. Retina. 2006;26:454–461.
171. Tatar O, Adam A, Shinoda K, et al. Matrix metalloproteinases in human choroidal neovascular
membranes excised following verteporfin photodynamic therapy. Br J Ophthalmol. 2007;91:1183–1189.
172. Beatty S, Koh H, Phil M, et al. The role of oxidative stress in the pathogenesis of age-related macular
degeneration. Surv Ophthalmol. 2000;45:115–134.
173. Jarrett SG, Boulton ME. Consequences of oxidative stress in age-related macular degeneration. Mol
Aspects Med. 2012;33:399–417.
174. Kaarniranta K, Elo M, Sironen R, et al. Hsp70 accumulation in chondrocytic cells exposed to high
continuous hydrostatic pressure coincides with mRNA stabilization rather than transcriptional activation.
Proc Natl Acad Sci U S A. 1998;95:2319–2324.
175. Kaarniranta K, Salminen A, Eskelinen E, et al. Heat shock proteins as gatekeepers of proteolytic
pathways—implications for age-related macular degeneration (AMD). Ageing Res Rev. 2009;8:128–139.
176. Kaarniranta K, Salminen A. Age-related macular degeneration: activation of innate immunity system
via pattern recognition receptors. J Mol Med. 2009;87:117–123.
177. Venza I, Visalli M, Oteri R, et al. Combined effects of cigarette smoking and alcohol consumption on
antioxidant/oxidant balance in age-related macular degeneration. Aging Clin Exp Res. 2012;24:530–536.
178. Hunter A, Spechler PA, Cwanger A, et al. DNA methylation is associated with altered gene expression
in AMD. Invest Ophthalmol Vis Sci. 2012;53:2089–2105.
179. Shaw PX, Zhang L, Zhang M, et al. Complement factor H genotypes impact risk of age-related
macular degeneration by interaction with oxidized phospholipids. Proc Natl Acad Sci U S A.
2012;109:13757–13762.
180. Nilsson I, Shibuya M, Wennstrom S. Differential activation of vascular genes by hypoxia in primary
endothelial cells. Exp Cell Res. 2004;299:471–485.
181. Hasty P, Campisi J, Hoeijimakers J, et al. Aging and genome maintenance: lessons from the mouse?.
Science. 2003;299:1355–1359.
182. Soussi T. The p53 tumor suppressor gene: from molecular biology to clinical investigation. Ann N Y
Acad Sci. 2000;910:121–137.
183. Singhal SS, Godley BF, Chandra A, et al. Induction of glutathione S transferase hGST 5.8 is an early
response to oxidative stress in RPE cells. Invest Ophthalmol Vis Sci. 1999;40:2652–2659.
184. Sasaki H, Ray PS, Zhu L, et al. Oxidative stress due to hypoxia/reoxygenation induces angiogenic
factor VEGF in adult rat myocardium: possible role of NFkappaB. Toxicology. 2000;30(155):27–35.
185. Duncan KG, Bailey KR, Kane JP, et al. Human retinal pigment epithelial cells express scavenger
receptors BI and BII. Biochem Biophys Res Commun. 2002;292:1017–1022.
186. Ryeom SW, Sparrow JR, Silverstein RL. CD36 participates in the phagocytosis of rod outer segments
by retinal pigment epithelium. J Cell Sci. 1996;109:387–395.
187. Armstrong D, Ueda TO, Ueda TA, et al. Lipid peroxide stimulates retinal neovascularization in rabbit
retina through expression of TNFa, VEGF and PDGF. Angiogenesis. 1998;2:174–184.
188. Terpstra V, Bird DA, Steinberg D. Evidence that the lipid moiety of oxidized low density lipoprotein
plays a role in its interaction with macrophage receptors. Proc Natl Acad Sci U S A.
1998;17(95):1806–1811.
189. Boullier A, Bird DA, Chang MK, et al. Scavenger receptors, oxidized LDL and atherosclerosis. Ann N
Y Acad Sci. 2001;947:214–222.
190. Ueda TO, Ueda TA, Fukuda S, et al. Lipid peroxide undiced TNFa, VEGF and neovascularization in
the rabbit cornea, effect of TNF inhibition. Angiogenesis. 1998;2:174–184.
191. Tamai K, Spaide RF, Ellis EA, et al. Lipid hydroperoxide stimulates subretinal choroidal
neovascularization in the rabbit. Exp Eye Res. 2002;74:301–308.
192. Honda S, Hjelmeland LM, Handa JT. Senescence associated beta galactosidase activity in human
retinal pigment epithelial cells exposed to mild hyperoxia in vitro. Br J Ophthalmol. 2002;86:159–162.
193. Zhang C, Baffi J, Cousins SW, et al. Oxidant induced cell death in retinal pigment epithelium cells
mediated through release of apoptosis-inducing factor. J Cell Sci. 2003;116:1915–1923.
5
ETIOLOGY OF LATE AGERELATED
MACULAR DISEASE
MARIANA INGOLOTTI • ERIC P. JABLON JOHN
BARNWELL KERRISON • HUGO QUIROZ-MERCADO D.
VIRGIL ALFARO III
INTRODUCTION
Age-related macular degeneration (AMD) is the leading cause of central vision
loss in the over-65 population in developed countries. It is a bilateral,
degenerative, and chronic disease limited to the macula so that the peripheral
retinal function is relatively preserved. Progressive histopathologic changes (Fig.
5.1) in the retinal pigment epithelium (RPE), retina photoreceptor cell layer,
Bruch's membrane (BM), and choriocapillaris are seen in these patients.
Figure 5.1 Histopathological changes in the two main forms of AMD. This diagram is showing the
structural and functional changes of a normal RPE cell when developing dry or wet AMD. A. The RPE in a
young retina has a homogenous distribution of the melanin granules, a BM without deposits, and a
preserved function. B. In dry AMD, the RPE melanin granules are localized basally, and lipofuscin
granules increase in number while the efficacy of the disks digestion reduces. Note the formation of drusen
between the basement membrane of the RPE and the inner collagen layer of the BM and the thickened BM.
C. In wet AMD, CNV develops, and an inflammatory response develops with an influx of lymphocytes
and macrophages to the scene. These fragile new vessels leak and lead to subretinal hemorrhage.
DRY/NONNEOVASCULAR AMD
Drusen
Drusen are deposits of extracellular matrix localized between the basement
membrane of the RPE and the inner collagen layer of the BM. They can extend
to the outer collagenous zone if there is discontinuity in the central band of
elastic fibers (10,11). They are often seen in the aging retina, as a hereditary
condition (dominantly inherited drusen) or secondary to a variety of intraocular
processes including inflammation, trauma, and chronic retinal detachment (2).
However, an increase in number, size, and confluence was established as a risk
factor for the development of AMD (12). Because of this, it is important to
distinguish which yellow deposits will lead to atrophy of the RPE or CNV. The
American Academy of Ophthalmology proposed the following classification
according to the size of the drusen (13):
Geographic Atrophy
Geographic or areolar atrophy also called advanced “dry” AMD involves an
extensive degeneration (150 μm or larger) of the RPE and the overlying
receptors that are metabolically dependent upon the RPE. It is labeled as
“geographic” because the areas of atrophic RPE are well demarcated and not
related to any specific anatomy structure (32). Some authors describe a
nongeographic atrophy in cases where atrophic areas are noncontiguous or
manifest as speckled depigmented areas (13). On funduscopic examination,
geographic atrophy is seen as a sharply demarcated round or oval
hypopigmented spot, frequently found either juxtafoveal or parafoveal, that
allows an enhanced view of the underlying large choroidal vessels (1,33). On
fluorescein angiography, geographic atrophy demonstrates an early
hyperfluorescent area due to the window defect in the RPE. The loss of
epithelium transmits fluorescein brightly, but the extent of the lesion does not
change in the late phases.
On microscopy, areas of geographic atrophy lack photoreceptors and RPE
cells, and the outer plexiform layer is adherent to the BM (Fig. 5.3A and B) (4).
This is seen as hypopigmented or atrophic areas overlying a diffusive thickened
inner aspect of the BM (26). Calcified or retractile drusen, as previously
described, are also observed (28). The RPE demonstrates other changes such as
hypertrophy and hyperplasia, which are clinically seen as focal
hyperpigmentation (23). Clumps of pigmented cells in the subretinal space and
the outer retinal layers may also be observed histologically (26).
Figure 5.3 Geographic atrophy. A. Retinal and choroidal tissue. Photoreceptor lesion with fibrosis (arrow)
H&E stain 20×. B. Macular retinal atrophy and fibrosis (arrow). Masson trichrome digital stain 60×.
(Courtesy Zárate JO, Alvarado M© 2011. Unpublished data.)
WET/EXUDATIVE/NEOVASCULAR AMD
Choroidal Neovascularization
Choroidal also known as subretinal neovascularization (CNV) is one of the main
features of wet AMD. The new vessel formation extends from the choroid into
the thickened portion of the BM or from the margins of the optic nerve head
(40). Studies have confirmed that a prior break in the BM is not necessary for
vessel proliferation because the new vessels can make their way through it by
themselves (41). CNV is often seen in retinas in which softening and confluence
of drusen have taken place (42). In histologic sections, Albert et al. (11)
described the following findings: (a) breaks in BM; (b) a granulomatous
inflammatory pattern with macrophages, lymphocytes, and fibroblasts; (c) basal
laminar deposits and soft drusen; and (d) RPE depigmentation, hypertrophy,
hyperplasia, and folding. On electron microscopy, both mature and newly
formed capillaries have fenestrations similar to their choriocapillaris precursors.
They also have high permeability to fluorescein and may cause leakage or
fluorescein blockage (43). Even though they are initially very small, are
nondetectable, and have capillary characteristics, they acquire arterial and
venous features with time. Ophthalmoscopically, they can either be seen through
the atrophic RPE or hidden underneath fibrous grayish tissue or RPE
hypertrophy. CNV can be localized to the macula, peripapillary retina, or
peripheral retina. CNV can be restricted to the BM (Fig. 5.4A and B) or trespass
the RPE and grow into the subsensory space associated with fibroblastic RPE
metaplasia and migration of macrophages (33). Anastomosis between choroidal
and retinal vessels can develop. This characteristic is often seen in late stages of
AMD when the disciform scar is well established (2). The neovascular
membranes can be recognized in the fluorescein angiogram as a fine network of
vessels with early filling, associated with exudation and subretinal hemorrhage.
These specific elements are toxic for the RPE and photoreceptors, and they may
lead to RPE detachment, proliferation of fibrous tissue, cystoid macular edema,
and occasional circinate retinopathy (33,44,45). In this setting, fibroblasts can be
identified invading the hematoma. Also, the surviving RPE cells are transformed
into elongated spindly cells while the overlying photoreceptor atrophy resulting
in a submacular nodule, plaque, or fibroglial scar tissue that contracts and
distorts the retina (40).
Figure 5.4 Wet AMD. A. Histologic section shows CNV at the BM. Retinal pigment epithelium (RPE),
choroid (C), and sclera (S). H&E stain 40×. B. Magnified view of the choroidal neovessels (asterisk). H&E
stain 60× (Courtesy Zárate JO, Alvarado M© 2011. Unpublished data.)
Disciform Scar
The formation of a disciform scar represents the end stage of AMD. As
previously described, the CNV fragile endothelium is associated with leakage
and bleeding, leading to RPE detachments. In the process of reorganizing the
damaged tissue, a fibrovascular disciform scar is generated.
The major histopathologic components of this scar are mounds of dense
collagenous connective tissue on the inner surface of the BM (24). It is generally
associated with the loss of neural tissue (photoreceptors) in the outer retina. As a
general rule, the thinner and smaller the scar's diameter, the greater the
photoreceptor survival (33). In spite of this, morphometric studies confirmed that
the ganglion cell and inner nuclear layers are relatively preserved (53). Cystoid
degeneration and lamellar and full-thickness macular holes are other examples of
possible degenerative changes (4). RPE proliferation, hyperplasia, and fibrous
metaplasia have been reported. These epithelial cells are responsible for the
production of large quantities of extracellular matrix material. The disciform
lesions can be partially pigmented because of melanin and hemosiderin presence
and may be difficult to differentiate from nevi, malignant melanoma, or pigment
epithelium tumors (45). Vascular supply is provided by choroidal vessel in most
cases, rarely by retinal vessels (4).
A rare but devastating complication of this condition is a massive subretinal
hemorrhage. It has been documented by Wood et al. (54) that neovascular tissue
present within the macular disciform lesions was the source of such bleedings
after a complete retinal detachment occurred. Other studies suggest that
anticoagulant medication, widely used among the AMD age group, can be a risk
factor for developing this unfortunate outcome. Therefore, the authors
recommend that anticoagulant therapy should be prescribed in cases only when
it is indicated (55). Moreover, another complication known as senile Coats'
response can also develop, resulting in extensive exudation in and under the
retina with lipid and cholesterol deposition (16).
REFERENCES
1. de Jong PT. Age-related macular degeneration. N Engl J Med. 2006;355(14):1474–1485.
2. Spencer WH; American Academy of Ophthalmology. Ophthalmic pathology: an atlas and textbook. 4th
ed. Philadelphia, PA: W.B. Saunders; 1996.
3. Weiter JJ, et al. Retinal pigment epithelial lipofuscin and melanin and choroidal melanin in human eyes.
Invest Ophthalmol Vis Sci. 1986;27(2):145–152.
4. Hampton GR, Nelsen PT. Age-related macular degeneration: principles and practice. New York: Raven
Press; 1992:300.
5. Huang JD, Curcio CA, Johnson M. Morphometric analysis of lipoprotein-like particle accumulation in
aging human macular Bruch's membrane. Invest Ophthalmol Vis Sci. 2008;49(6):2721–2727.
6. Huang JD, et al. Age-related changes in human macular Bruch's membrane as seen by quick-
freeze/deep-etch. Exp Eye Res. 2007;85(2):202–218.
7. Curcio CA, et al. Accumulation of cholesterol with age in human Bruch's membrane. Invest Ophthalmol
Vis Sci. 2001;42(1):265–274.
8. Booij JC, et al. The dynamic nature of Bruch's membrane. Prog Retin Eye Res. 2010;29(1):1–18.
9. Shelley EJ, et al. Cone degeneration in aging and age-related macular degeneration. Arch Ophthalmol.
2009;127(4):483–492.
10. Farkas TG, Sylvester V, Archer D. The ultrastructure of drusen. Am J Ophthalmol.
1971;71(6):1196–1205.
11. Albert DM, Jakobiec FA. Principles and practice of ophthalmology. 3rd Ed. Philadelphia, PA: Elsevier
Saunders; 2007.
12. Bressler SB, et al. Relationship of drusen and abnormalities of the retinal pigment epithelium to the
prognosis of neovascular macular degeneration. The Macular Photocoagulation Study Group. Arch
Ophthalmol. 1990;108(10):1442–1447.
13. Regillo CD, Holekamp N, Johnson MW, et al. Basic and clinical science course section 12. Retina and
vitreous 2008-2009. San Francisco, CA:The Foundation of the American Academy of Ophthalmology;
2008:124–136.
14. Nowak JZ. Age-related macular degeneration (AMD): pathogenesis and therapy. Pharmacol Rep.
2006;58(3):353–363.
15. Schmack I, Kang SJ, Grossniklaus HE. Histopatologia de la degeneracion macular asociada a la edad.
Mones J, Gomez-Ulla F, eds. Degeneracion macular associada a la edad. Barcelona: Proust Science;
2005:73–84.
16. Green WR, McDonnell PJ, Yeo JH. Pathologic features of senile macular degeneration.
Ophthalmology. 1985;92(5):615–627.
17. Ulshafer RJ, et al. Scanning electron microscopy of human drusen. Invest Ophthalmol Vis Sci.
1987;28(4):683–689.
18. Burns RP, Feeney-Burns L. Clinico-morphologic correlations of drusen of Bruch's membrane. Trans
Am Ophthalmol Soc. 1980;78:206–225.
19. Hogan MJ. Role of the retinal pigment epithelium in macular disease. Trans Am Acad Ophthalmol
Otolaryngol. 1972;76(1):64–80.
20. Fine BS. Lipoidal degeneration of the retinal pigment epithelium. Am J Ophthalmol.
1981;91(4):469–473.
21. el Baba F, et al. Clinicopathologic correlation of lipidization and detachment of the retinal pigment
epithelium. Am J Ophthalmol. 1986;101(5):576–583.
22. Sassani JW. Ophthalmic pathology with clinical correlations. Philadelphia, PA: Lippincott-Raven;
1997:337.
23. Green WR. Histopathology of age-related macular degeneration. Mol Vis. 1999;5:27.
24. Eagle RC. Eye pathology: an atlas and basic text. Philadelphia, PA: W.B. Saunders; 1999:306.
25. Lommatzsch A, et al. Are low inflammatory reactions involved in exudative age-related macular
degeneration? Morphological and immunohistochemical analysis of AMD associated with basal deposits.
Graefes Arch Clin Exp Ophthalmol. 2008;246(6):803–810.
26. Bressler NM, et al. Clinicopathologic correlation of drusen and retinal pigment epithelial abnormalities
in age-related macular degeneration. Retina. 1994;14(2):130–142.
27. Sarks JP, Sarks SH, Killingsworth MC. Evolution of geographic atrophy of the retinal pigment
epithelium. Eye (Lond). 1988;2(Pt 5):552–577.
28. Cukras C, et al. Natural history of drusenoid pigment epithelial detachment in age-related macular
degeneration: Age-Related Eye Disease Study Report No. 28. Ophthalmology. 2010;117(3):489–499.
29. Zarbin MA, Age-related macular degeneration: review of pathogenesis. Eur J Ophthalmol.
1998;8(4):199–206.
30. Jager RD, Mieler WF, Miller JW. Age-related macular degeneration. N Engl J Med.
2008;358(24):2606–2617.
31. Kenyon KR, et al. Diffuse drusen and associated complications. Am J Ophthalmol.
1985;100(1):119–128.
32. Schatz H, McDonald HR. Atrophic macular degeneration. Rate of spread of geographic atrophy and
visual loss. Ophthalmology. 1989;96(10):1541–1551.
33. Hamilton AMP, Gregson R, Fish GE. Text atlas of the retina. Boston, Oxford: Butterworth-Heinemann;
Martin Dunitz; 1998:417.
34. Liu X, et al. Extracellular matrix of retinal pigment epithelium regulates choriocapillaris endothelial
survival in vitro. Exp Eye Res. 1997;65(1): 117–126.
35. Ciulla TA, et al. Color Doppler imaging discloses reduced ocular blood flow velocities in nonexudative
age-related macular degeneration. Am J Ophthalmol. 1999;128(1):75–80.
36. Kliffen M, et al. Morphologic changes in age-related maculopathy. Microsc Res Tech.
1997;36(2):106–122.
37. Hoskin A, Bird AC, Sehmi K. Tears of detached retinal pigment epithelium. Br J Ophthalmol.
1981;65(6):417–422.
38. Gass JD. Pathogenesis of tears of the retinal pigment epithelium. Br J Ophthalmol.
1984;68(8):513–519.
39. Gelisken F, et al. Retinal pigment epithelial tears after single administration of intravitreal bevacizumab
for neovascular age-related macular degeneration. Eye (Lond). 2009;23(3):694–702.
40. Lucas DR, Greer CH. Greer's ocular pathology. 4th Ed. Oxford, Boston: Blackwell Scientific
Publications; 1989:339.
41. Heriot WJ, et al. Choroidal neovascularization can digest Bruch's membrane. A prior break is not
essential. Ophthalmology. 1984;91(12):1603–1608.
42. Sarks SH. Council Lecture. Drusen and their relationship to senile macular degeneration. Aust J
Ophthalmol. 1980;8(2):117–130.
43. Miller H, Miller B, Ryan SJ. Newly-formed subretinal vessels. Fine structure and fluorescein leakage.
Invest Ophthalmol Vis Sci. 1986;27(2):204–213.
44. Green WR, Key SN III. Senile macular degeneration: a histopathologic study. Trans Am Ophthalmol
Soc. 1977;75:180–254.
45. Apple DJ, Rabb MF. Ocular pathology: clinical applications and self-assessment. 5th Ed. St. Louis,
MO: Mosby; 1998:705.
46. Grossniklaus HE, Gass JD. Clinicopathologic correlations of surgically excised type 1 and type 2
submacular choroidal neovascular membranes. Am J Ophthalmol. 1998;126(1):59–69.
47. Yannuzzi LA, et al. Retinal angiomatous proliferation in age-related macular degeneration. Retina.
2001;21(5):416–434.
48. Hartnett ME, et al. Classification of retinal pigment epithelial detachments associated with drusen.
Graefes Arch Clin Exp Ophthalmol. 1992;230(1):11–19.
49. Slakter JS, et al. Retinal choroidal anastomoses and occult choroidal neovascularization in age-related
macular degeneration. Ophthalmology. 2000;107(4):742–753; discussion 753–754.
50. Lafaut BA, et al. Clinicopathological correlation of deep retinal vascular anomalous complex in age
related macular degeneration. Br J Ophthalmol. 2000;84(11):1269–1274.
51. Yannuzzi LA, Freund KB, Takahashi BS. Review of retinal angiomatous proliferation or type 3
neovascularization. Retina. 2008;28(3):375–384.
52. Shimada H, et al. Clinicopathological findings of retinal angiomatous proliferation. Graefes Arch Clin
Exp Ophthalmol. 2007;245(2):295–300.
53. Kim SY, et al. Morphometric analysis of the macula in eyes with geographic atrophy due to age-related
macular degeneration. Retina. 2002;22(4):464–470.
54. Wood WJ, Smith TR. Senile disciform macular degeneration complicated by massive hemorrhagic
retinal detachment and angle closure glaucoma. Retina. 1983;3(4):296–303.
55. Tilanus MA, et al. Relationship between anticoagulant medication and massive intraocular hemorrhage
in age-related macular degeneration. Graefes Arch Clin Exp Ophthalmol. 2000;238(6):482–485.
SECTION II
INTRODUCTION
Age-related macular degeneration (AMD) has been subject to many divisions,
namings, and classification schemes since it was first described by Haab in 1885
(1). The disease has since been subdivided into two major forms: nonexudative
(dry AMD) and exudative (wet AMD). In nonexudative AMD, visual loss
develops as the result of geographic atrophy involving the foveal center (2,3). In
exudative AMD, visual loss develops secondary to choroidal neovascularization
(CNV), which leads to bleeding, exudation, and eventual scar formation (4).
Exudative AMD is distinguished from nonexudative AMD when the integrity of
the Bruch's membrane–retinal pigment epithelium (RPE) complex separates and
forms a pigment epithelial detachment (PED). While nonexudative AMD has not
been subject to subdivisions, exudative AMD has several subclassifications. In
the past, exudative AMD has been most commonly divided into occult and
classic types, based on characteristic lesion formation. In classic exudative
AMD, lesion boundaries were well demarcated on fluorescein angiography (FA),
while occult lesions had irregular boundaries and stippled hyperfluorescence.
Other classifications have been based on the grading systems of various studies
and clinical trials (5). In 1995, based on postoperative results in patients with
CNV, Gass (6,7) suggested a shift in the classification of exudative AMD using
the anatomic localization rather than the appearance on FA as was proposed in
the Macular Photocoagulation Study (MPS) (8,9). Such classification was
limited by the lack of imaging available at the time but can now be used due to
multimodal imaging, employing the use of FA, optical coherence tomography
(OCT), and indocyanine green (ICG) angiography.
A new classification scheme proposed by Freund et al. (10) builds on the
Gass model, dividing neovascularization into four types depending on location:
type 1, located under the RPE; type 2, above the RPE, in the subretinal space;
type 3, characterized by intraretinal neovascularization (IRN), also known as
retinal angiomatous proliferation (RAP); and type 4, polypoidal choroidal
vasculopathy (PCV), considered by some to be type 1 neovascularization. While
PCV is a sub-RPE vascular lesion, it does not have the clinical, structural, and
angiographic characteristics of type 1 lesions. This method of classification
would not rely on only one imaging technology as in the past with FA but also
on imaging techniques that emerged in recent years, refining our understanding
of the disease.
TYPE 1 NEOVASCULARIZATION
Gass defined type 1 neovascularization histologically as neovascularization
occurring beneath the RPE monolayer. It is by far the most common form of
neovascularization in AMD. In a study by Cohen et al. (11), occult CNV with or
without PED represented 56.6% of the 207 cases studied. With FA, these vessels
are usually described as occult or poorly defined. In a study by Lafaut et al. (12),
occult membranes were analyzed after surgical excision. All cases consisted of
thin fibrovascular membranes in the sub-RPE space. Only in two of the ten
specimens was an additional smaller fibrovascular component found in the
subretinal space.
FA terminology applied to type 1 lesions includes “late leakage of
undetermined source” or vascularized PED. ICG was found to be useful in
detecting choroidal neovascular membranes (CNVMs), especially if FA cannot
reveal a well-defined neovascular membrane (Figs. 6.1A and 6.2A). ICG is a
complementary imaging study in the diagnosis of occult lesions where masking
phenomena may prevent the visualization of the neovascular net. ICG is less
dependent on leakage as ICG dye bound to albumin is kept in the intravascular
space (13,14). This relatively new technique helped in deciding the eligibility of
treatment with photodynamic therapy (PDT) in some occult CNV cases (15).
Yannuzzi and Slakter (16,17) found that 39% and 44%, respectively, of patients
with occult CNV by FA showed well-demarcated hyperfluorescence by ICG
angiography. The ICG pattern of type 1 neovascularization can be classified into
three types depending on the size of the hyperfluorescent area: focal spots,
plaques, or a combination of both (18). In a postmortem histopathologic study of
an eye with ICG plaque, Chang et al. (19) confirmed the localization of plaques
documented by ICG to be between the RPE and an intact Bruch's membrane.
FIGURE 6.1 A. Type 1 CNVM. Left. FA showing leakage of undetermined origin. Right. ICG
angiography showing arteriolized pattern of choroidal neovascularization. B.Type 1 CNVM. Left. ICG
angiography showing arteriolized pattern of CNV. Right. Macular OCT showing a visible large pigment
epithelium detachment (PED).
FIGURE 6.2 A. Type 1 CNVM. Early- and late-phase FA: Leakage of undetermined origin. PED is
visible. B.Type 1 CNVM. Left. ICG: Bushy appearance of CNVM apparent with ICG. Right. OCT of the
same case. Pigment epithelium detachment with minimal subretinal fluid.
TYPE 2 NEOVASCULARIZATION
As mentioned previously, the type 2 pattern predominates in macular disorders
where there is a focal alteration of the RPE–Bruch's membrane complex such as
pathologic myopia with lacquer cracks (Fig. 6.3A and B), punctate inner
choroidopathy, multifocal choroiditis and panuveitis, histoplasmosis, and
choroidal rupture. It is seen also in entities where material is deposited above the
RPE such as vitelliform macular dystrophies, Best's disease, Stargardt's disease,
subretinal drusenoid deposits (reticular pseudodrusen), and pseudoxanthoma
elasticum (10).
FIGURE 6.3 A. Type 2 membrane in a case with high myopia. FA (early and late phases) showing leak in
a classic pattern. B. OCT of the same case. Disruption of the RPE is observed as well as a hyperreflective
area anterior to the RPE that corresponds to a type 2 CNVM.
On OCT, a high level of correlation was found between areas of classic CNV
seen on FA and the volume of subretinal tissue (32). It is possible to differentiate
a classic CNV using SD-OCT. The retinal thickness map specifically shows a
well-defined lesion with steep borders and a craterlike central depression (14).
Leakage and staining can be easily differentiated with this imaging technique
where hyperreflectivity represents visualization of fibrotic tissue and not edema.
With OCT, type 2 membranes can be localized above the RPE and beneath the
photoreceptor outer segments (10) (Fig. 6.5B).
The hyperreflective line, anterior to the RPE line, represents the inner
segment–outer segment junction of the photoreceptors (33). Disorganization of
this layer, overlying the inner segment–outer segment (IS/OS) junction, is often
accompanied by intraretinal cystic spaces. Intraretinal fluid rather than subretinal
fluid predominates in type 2 lesions and usually is associated with decreased
visual acuity (34,35). The differentiation between these two types of leakage can
be easily seen with OCT but is not possible with FA (10).
In patients with classic lesions, most vision loss occurs within the first 6
months with little vision loss after the first year. Predominantly classic lesions
included in the TAP trial were found to be smaller in size than the occult (type 1)
lesions included in the VIP trial. It is suggested that predominantly classic
lesions almost always cause visual symptoms promptly, perhaps allowing
identification of smaller lesion sizes, whereas occult lesions may or may not
cause prompt visual symptoms, which allows them to grow to larger sizes
(23,28).
Early in their evolution, type 2 vessels appear to be exquisitely sensitive to
intravitreal anti-VEGF agents. There may be a decrease in the volume of
subretinal fluid and fibrovascular tissue. In larger and more mature lesions, the
abnormal vessels remain as a hyperreflective band (subretinal fibrosis) causing
disorganization and thinning of the overlying receptors. This is seen as a
disruption of the IS/OS junction, loss of the ELM, and thinning of the outer
nuclear layer on SD-OCT. These eyes may continue to manifest intraretinal fluid
despite frequent treatment, presumably due to the loss of the outer blood–retinal
barrier in the form of RPE damage and disruption of the tight junctions that
contribute to ELM band on the OCT (10). Even in eyes with early lesions, the
disruption of the IS/OS layer may be indicative of the future visual result, and
visual recovery may be limited in these eyes because of the initial assault to the
photoreceptors (33).