ArticlePDF Available

The RAGE axis in early diabetic retinopathy

Authors:
  • Astellas Institute for Regenerative Medicine

Abstract and Figures

The receptor for advanced glycation end products (AGEs) has been implicated in the pathogenesis of diabetic complications. This study was conducted to characterize the role of the RAGE axis in a murine model of nonproliferative diabetic retinopathy (NPDR). The retinas of hyperglycemic, hyperlipidemic (HGHL, apolipoprotein E(-/-) db/db) mice were examined for the development of early retinal vascular lesions of NPDR and compared to littermates at 6 months of age. Neural function was assessed with electroretinography. Immunohistochemistry, real-time RT-PCR, autofluorescence, and ELISA studies were used to localize and quantify the AGE/RAGE axis. Soluble RAGE, a competitor of cellular RAGE for its ligands, was administered to assess the impact of RAGE blockade. Early inner retinal neuronal dysfunction, manifested by prolonged latencies of the oscillatory potentials and b-wave, was detected in hyperglycemic mice. HGHL mice exhibited accelerated development of acellular capillaries and pericyte ghosts compared with littermate control animals. AGEs were localized primarily to the vitreous cavity and internal limiting membrane (ILM) of the retina, where they were intimately associated with the footplates of RAGE-expressing Müller cells. AGE accumulation measured by ELISA was increased within the retinal extracellular matrix of hyperglycemic mice. AGE fluorescence and upregulation of RAGE transcripts was highest in the retinas of HGHL mice, and attenuation of the RAGE axis with soluble RAGE ameliorated neuronal dysfunction and reduced the development of capillary lesions in these mice. In early diabetic retinopathy, the RAGE axis, comprising the cellular receptor and its AGE ligands, is amplified within the retina and is accentuated along the vitreoretinal interface. Antagonism of the RAGE axis in NPDR reduces neurovascular perturbations, providing an important therapeutic target for intervention.
Content may be subject to copyright.
The RAGE Axis in Early Diabetic Retinopathy
Gaetano R. Barile,
1
Sophia I. Pachydaki,
1
Samir R. Tari,
1
Song E. Lee,
1
Christine M. Donmoyer,
1
Wanchao Ma,
1
Ling Ling Rong,
2
Loredana G. Buciarelli,
2
Thoralf Wendt,
2
Heidi Ho¨rig,
2
Barry I. Hudson,
2
Wu Qu,
2
Alan D. Weinberg,
3
Shi Fang Yan,
2
and Ann Marie Schmidt
2
PURPOSE. The receptor for advanced glycation end products
(AGEs) has been implicated in the pathogenesis of diabetic
complications. This study was conducted to characterize the
role of the RAGE axis in a murine model of nonproliferative
diabetic retinopathy (NPDR).
M
ETHODS. The retinas of hyperglycemic, hyperlipidemic
(HGHL, apolipoprotein E
/
db/db) mice were examined for
the development of early retinal vascular lesions of NPDR and
compared to littermates at 6 months of age. Neural function
was assessed with electroretinography. Immunohistochemis-
try, real-time RT-PCR, autofluorescence, and ELISA studies
were used to localize and quantify the AGE/RAGE axis. Soluble
RAGE, a competitor of cellular RAGE for its ligands, was ad-
ministered to assess the impact of RAGE blockade.
R
ESULTS. Early inner retinal neuronal dysfunction, manifested
by prolonged latencies of the oscillatory potentials and b-wave,
was detected in hyperglycemic mice. HGHL mice exhibited
accelerated development of acellular capillaries and pericyte
ghosts compared with littermate control animals. AGEs were
localized primarily to the vitreous cavity and internal limiting
membrane (ILM) of the retina, where they were intimately
associated with the footplates of RAGE-expressing Mu¨ller cells.
AGE accumulation measured by ELISA was increased within
the retinal extracellular matrix of hyperglycemic mice. AGE
fluorescence and upregulation of RAGE transcripts was highest
in the retinas of HGHL mice, and attenuation of the RAGE axis
with soluble RAGE ameliorated neuronal dysfunction and re-
duced the development of capillary lesions in these mice.
C
ONCLUSIONS. In early diabetic retinopathy, the RAGE axis,
comprising the cellular receptor and its AGE ligands, is ampli-
fied within the retina and is accentuated along the vitreoretinal
interface. Antagonism of the RAGE axis in NPDR reduces neu-
rovascular perturbations, providing an important therapeutic
target for intervention. (Invest Ophthalmol Vis Sci. 2005;46:
2916–2924) DOI:10.1167/iovs.04-1409
D
iabetic retinopathy, the leading cause of irreversible blind-
ness in the working population in the Western world,
encompasses both vascular and neural dysfunction.
1
Diabetes
mellitus leads to alterations in the perfusion and permeability
of the retinal vasculature, resulting in retinal ischemia and/or
edema, with loss of reading vision when these events occur in
the central macular region.
2
Diabetic retinopathy is also a
degenerative disease of the neural retina, associated with alter-
ations in neuronal function before the onset of clinical vascular
disease.
3
In advanced, proliferative diabetic retinopathy, an
angiogenic, VEGF-mediated response with retinal neovascular-
ization ensues, placing the eye at further risk of severe visual
loss due to the development of vitreous hemorrhage or trac-
tion retinal detachment.
4
Although many cases of diabetic
retinopathy may be amenable to treatment with laser photo-
coagulation or vitrectomy, such efforts may not prevent irre-
versible vascular or neuronal damage, thereby underscoring
the need for early intervention.
The duration and severity of hyperglycemia is the single
most important factor linked to the development of diabetic
retinopathy. The degree of hyperglycemia is the major alter-
able risk factor for both the development and progression of
diabetic retinopathy, in both types 1 and 2 diabetes, as seen in
the Diabetes Control and Complications Trial (DCCT)
5
and in
the UK Prospective Diabetes Study (UKPDS),
6
respectively.
Additional established risk factors for the acceleration of dia-
betic retinopathy include hypertension and hyperlipidemia,
with several clinical studies demonstrating benefit in the treat-
ment of diabetic retinopathy with intensive blood pressure
control and lipid-lowering therapy.
7–13
One metabolic conse-
quence of chronic hyperglycemia is the accelerated formation
of advanced glycation end products (AGEs), the accumulation
of which in diabetic tissues is enhanced not only by elevated
glucose but also by oxidant stress and inflammatory stimuli.
14
In the setting of diabetic retinopathy, AGEs, especially N
-
(carboxymethyl)lysine (CML) adducts, have been detected
within retinal vasculature and neurosensory tissue of diabetic
eyes.
15
Multiple consequences of AGE accumulation in the
retina have been demonstrated, including upregulation of
VEGF, upregulation of NF-
B, and increased leukocyte adhe-
sion in retinal microvascular endothelial cells.
16–18
In diabetic
patients, AGEs also accumulate within the vitreous cavity and
may result in characteristic structural alterations sometimes
referred to as diabetic vitreopathy.
19,20
Support for a role for
AGEs as a contributing factor in the pathogenesis of diabetic
retinopathy has been drawn from studies in animals with in-
hibitors of AGE formation.
21,22
In a 5-year study in diabetic
dogs, administration of aminoguanidine prevented retinopa-
thy. Similar beneficial effects in the retinal vasculature of dia-
betic rats have been observed with other inhibitors of AGE
formation, including pyridoxamine and benfotiamine.
23,24
AGEs exert cell-mediated effects via RAGE, a multiligand
signal-transduction receptor of the immunoglobulin superfam-
ily.
25
Coinciding with pathologic changes in tissues, RAGE
From the Departments of
1
Ophthalmology and
2
Surgery, College
of Physicians and Surgeons, and the
3
Department of Biostatistics,
Mailman School of Public Health, Columbia University, New York,
New York.
Supported by the Juvenile Diabetes Research Foundation, the
Columbia Irving Center for Clinical Research, the Eye Surgery Fund,
the Surgical Research Fund, and Research to Prevent Blindness. GRB is
a recipient of the Florence and Herbert Irving Clinical Research Scholar
Award and the Glaubinger Scholar Award. AMS is a recipient of the
Burroughs Wellcome Fund Clinical Scientist Award in Translational
Research.
Submitted for publication December 2, 2004; revised March 10,
2005; accepted March 29, 2005.
Disclosure: G.R. Barile, None; S.I. Pachydaki, None; S.R. Tari,
None; S.E. Lee, None; C.M. Donmoyer, None; W. Ma, None; L.L.
Rong, None; L.G. Buciarelli, None; T. Wendt, None; H. Ho¨rig,
None; B.I. Hudson, None; W. Qu, None; A.D. Weinberg, None; S.F.
Yan, None; A.M. Schmidt, None
The publication costs of this article were defrayed in part by page
charge payment. This article must therefore be marked advertise-
ment in accordance with 18 U.S.C. §1734 solely to indicate this fact.
Corresponding author: Gaetano R. Barile, Harkness Eye Institute,
635 West 165th Street, Box 94, New York, New York 10032;
grb17@columbia.edu.
Investigative Ophthalmology & Visual Science, August 2005, Vol. 46, No. 8
2916
Copyright © Association for Research in Vision and Ophthalmology
expression increases dramatically, with AGE ligands further
upregulating receptor expression to magnify local cellular re-
sponses.
26
RAGE also binds the proinflammatory mediators,
the S100/calgranulins, and amphoterin,
27,28
and is an endothe
-
lial cell adhesion receptor capable of promoting leukocyte
recruitment through interaction with the integrin Mac-1.
29
Consequences of ligand-RAGE interaction include increased
expression of vascular cell adhesion molecule (VCAM)-1, vas-
cular hyperpermeability, enhanced thrombogenicity, induc-
tion of oxidant stress and abnormal expression of eNOS, all
pathogenetic mechanisms that potentially contribute to the
ischemic and vasopermeability events of diabetic retinopa-
thy.
30,31
Based on these considerations, we examined the RAGE axis
in a newly characterized murine model of nonproliferative
diabetic retinopathy. We first bred hyperlipidemic apoE
/
mice into the hyperglycemic db/db background, observing
that hyperlipidemia accelerates structural vascular changes in
diabetic retinas that exhibit neuronal dysfunction. We localized
and quantified the RAGE axis—specifically, AGE ligands and
their cellular receptor RAGE, in the eyes of these mice. The
findings provide new insights into the role of the RAGE axis in
the pathogenesis of early diabetic retinopathy.
METHODS
Generation of the Mouse Colony
To generate the apoE
/
db/db mice, apoE
/
mice were first back
-
crossed six generations into mice heterozygous for the diabetes spon-
taneous mutation (Lepr
db
). As the homozygous db/db mouse is sterile,
we ultimately bred apoE
/
db/m offspring to generate apoE
/
db/db mice. Initially, male mice heterozygous for the diabetes sponta-
neous mutation (Lepr
db
) in the leptin receptor gene on chromosome
4 (BKS.Cg-m
/
Lepr
db
, former name C57BLK/J-m
/
Lepr
db
, Type
JAX GEMM TM Strain; Spontaneous Mutation Congenic, stock no.
000642; Jackson Laboratory, Bar Harbor, ME) were crossed with fe-
male mice homozygous for the Apoe
tm1Unc
mutation in chromosome 7
(B6.129P2-Apoe
tm1Unc
, former name C57BL/6J-Apoe
tm1Unc
, Type JAX
GEMM TM Strain; Targeted Mutation Congenic, stock no. 002052;
Jackson Laboratory) at approximately 8 weeks of age. All mice were
fed normal rodent chow (5053; PMI Nutrition International, Inc., St.
Louis, MO) and exposed to a 12-hour light-dark cycle. All offspring
were heterozygous for the apoE mutation. The genotype of their
offspring was identified by PCR with primers from Invitrogen Corp.
(Carlsbad, CA). The heterozygous mice from different parents were
again crossed at 8 weeks of age. Mice homozygous for the Apoe
tm1Unc
mutation and heterozygous for the Lepr
db
mutation (apoE
/
db/m)
were used as breeders and were crossed with one another to breed the
double-knock-out apoE
/
db/db mice. Control mice were littermates
obtained from the same colony: apoE
/
db/m mice (homozygous for
the wild type allele Apoe
tm1Unc
and heterozygous for the db mutation)
which are normoglycemic, nonobese littermates; apoE
/
db/db mice
(homozygous for the wild-type allele Apoe
tm1Unc
and homozygous for
the Lepr
db
mutation) which are hyperglycemic, normolipidemic litter
-
mates. Glucose measurements were performed during the course of
generation of the colony with a glucometer (Freestyle; Therasense,
Alameda, CA). Cholesterol measurements were then performed (Infin-
ity Cholesterol Liquid Stable Reagent kit; Thermo Electron Corp.,
Waltham, MA). The generation of the colony and all experiments were
performed in agreement with the ARVO Statement for the Use of
Animals in Ophthalmic and Vision Research and were approved by the
Institutional Animal Care and Use Committee at Columbia University.
Elastase Retinal Digest
An elastase digest with histopathologic vascular analysis was per-
formed on 35 mice at age 6 months, including analysis of the following
phenotypes: apoE
/
db/m (n 7; normoglycemic, normolipidemic
[NGNL]); apoE
/
db/m (n 8; normoglycemic, hyperlipidemic
[NGHL]); apoE
/
db/db (n 7; hyperglycemic, normolipidemic
[HGNL]); and apoE
/
db/db (n 13; hyperglycemic, hyperlipidemic
[HGHL]). At the time of death, the eyes were enucleated and placed in
10% formalin for 2 days. After fixation, the retina was gently dissected
from the neurosensory retina under microscopic observation. The
neurosensory retina was placed in distilled water overnight to remove
fixative. The elastase digestion method described by Laver et al.
32
was
then performed. After the vascular specimen was mounted on a slide,
periodic acid-Schiff and hematoxylin staining of the vascular network
and nuclei was performed. The specimens were then analyzed by
microscope with digital capture (Axioskop 2 Plus; Carl Zeiss MicroIm-
aging Inc., Thornwood, NY) for the presence of acellular capillaries
and pericyte ghosts. Acellular capillaries were at least one-third thick-
ness of normal capillary width, and intercapillary bridges were ex-
cluded from analysis.
33
The examiner was masked to the nature of the
specimen during the assessment of pathology. As vascular lesions may
be distributed nonuniformly, the entire retina was scanned during this
process, and images were pasted into a single image (Photoshop, ver.
7.0; Adobe Systems Inc., San Jose, CA) to obtain an image of wholem-
ounted retina for area calculations. The virtual area of each prepared
retina was measured (OphthaVision Imaging System, ver. 3.25; MRP
Group Inc., Lawrence, MA). The number of acellular capillaries and
pericyte ghosts for each digest was divided by the area scanned. The
data obtained were analyzed with frequency and descriptive statistics,
as described below.
Electrophysiology
Electroretinograms (ERGs) were performed on the following age-
matched, 6-month-old littermates: NGNL wild-type (apoE
/
db/m; n
18), NGHL (apo E
/
db/m; n 11), HGNL (apoE
/
db/db; n
8), and HGHL (apoE
/
db/db; n 14) mice. The mice were dark
adapted overnight before each experiment, and the ensuing proce-
dures were performed under dim red light in a darkroom. The mice
were anesthetized with a mixture of 50 mg/kg ketamine and 5 mg/kg
xylazine administered intraperitoneally. The right eye pupil was dilated
with drops of 2.5% phenylephrine hydrochloride and 0.5% tropicam-
ide. The electroretinogram (ERG) responses were amplified and aver-
aged by a computerized data-acquisition system (PowerLab; ADInstru-
ments, Colorado Springs, CO). Once anesthetized, the mouse was
placed on a heated block, and body temperature was maintained near
37°C. The mouse was placed in a centered position at the edge of a
Ganzfeld dome. A rectal thermometer was placed in the mouse and
checked throughout the recording. A ground electrode was inserted in
the right leg, and the reference electrode was inserted in the forehead.
The data collected and analyzed included all the above and tempera-
ture of the animal during the experiment, a- and b-wave latency and
amplitude, oscillatory potential (OP)1, OP2, and OP3 implicit times
and amplitudes, as previously described.
34,35
The data obtained were
analyzed with frequency and descriptive statistics, as described later.
Immunochemical Staining
Eyes of 6-month-old mice were fixed overnight in 4% phosphate-
buffered paraformaldehyde and embedded in paraffin. The 4-mm par-
affin sections were deparaffinized and heated in citrate buffer with a
microwave for 15 minutes. After pretreatment with PBS containing 5%
normal goat serum (Jackson ImmunoResearch Laboratories Inc., West
Grove, PA), 0.5% BSA, and 0.1% Triton X-100 for 30 minutes at room
temperature (RT), sections were incubated with anti-mouse RAGE
antibody
36
(1:100), anti-AGE antibody
36
(1:100), anti-vimentin anti
-
body (1:200, Santa Cruz Biotechnology Inc., Santa Cruz, CA), anti-glial
fibrillary acidic protein (GFAP) antibody (1:100, Chemicon Interna-
tional, Inc., Temecula, CA), or anti-CD31 antibody (1:200, BD-Pharmin-
gen, San Diego, CA) for 1 hour at RT and then overnight at 4°C. After
they were rinsed with PBS, the sections were incubated for 1 hour at
RT with secondary antibody conjugated to Alexa Fluor 488 (Molecular
Probes Inc., Eugene, OR) or Alexa Fluor 546 (Molecular Probes Inc.).
All antibodies were diluted in PBS containing 0.5% goat serum, 0.5%
BSA, and 0.1% Triton X-100. Rabbit or chicken serum was used instead
IOVS, August 2005, Vol. 46, No. 8 RAGE in Early Diabetic Retinopathy 2917
of primary antibody for a negative control. The retina was examined
with a microscope (Eclipse E800; Nikon Instruments Inc., Meville, NY)
equipped with a confocal laser scanning system (Radiance2000; Bio-
Rad Laboratories, Hercules, CA). Images were captured and processed
(LaserSharp 2000 software; Bio-Rad Laboratories).
Autofluorescence and ELISA of Retinal AGEs
Five mice from each group were euthanatized. Whole retinas were
homogenized in 0.1 mL of PBS with 0.1% Triton X-100 at 0°C. Samples
were centrifuged at 20,000g for 5 minutes at 4°C. Protein concentra-
tion was determined with BSA used as a standard. The protein level in
the supernatant was adjusted to 1.6 mg/mL and used for a cellular
protein autofluorescence assay. The pellet, mostly extracellular matrix
(ECM), was washed with 20 mM phosphate buffer (pH 7.0), with 10
mM EDTA, and digested with 20
L of 25 U/mL papain (P5306,
Sigma-Aldrich, St. Louis, MO) in 20 mM phosphate buffer (pH 7.0) 10
mM EDTA, and 20 mM cysteine at 37°C. After 24 hours, another 20
L
of papain solution was added, and the incubation was continued for 24
hours. The supernatant was used for the measurement of ECM
autofluorescence and ELISA of AGEs after appropriate dilution. Fluo-
rescence intensities were measured on a multiwell plate reader (Cyto-
Fluor 4000; Applied Biosystems [ABI], Foster City, CA) using 360
40/460 40-nm excitation/emission wavelengths. These excitation/
emission wavelengths allow for detection of well-defined AGEs.
37,38
Fluorescence was expressed in fluorescence intensity per 0.1 mg
cellular protein or its equivalent retinal size for ECM. For immuno-
chemical measurement of AGEs in ECM, a noncompetitive ELISA was
used. The wells (96-well Nunc-Immuno Plate; Nalge Nunc Interna-
tional, Rochester, NY) were coated with BSA control, AGE-BSA stan-
dard,
36
and biological samples in 0.1 mL of 50 mM carbonate buffer
(pH 9.6) at 4°C overnight. The wells were then washed with PBS
containing 0.05% Tween-20 (washing buffer) and blocked at room
temperature with 0.3 mL of 1% BSA and 5% rabbit serum in PBS
(blocking buffer) for 1 hour. After they were washed, the wells were
incubated with anti-AGE antibody
36
in blocking buffer for 3 hours at
room temperature, followed by washing and secondary antibody (rab-
bit anti-chicken IgY-HRP; Biomeda Corp, Foster City, CA) for 1 hour at
room temperature. The wells were then washed again and developed
with 0.1 mL of peroxidase substrates (o-phenylenediamine tablets;
Sigma-Aldrich) in the dark at room temperature. The absorbance at 490
nm was measured after adding 0.05 mL of blocking solution (2 M
H
2
SO
4
) at 10 minutes.
Quantitative Real-Time PCR
At least five mice of each group were euthanatized. Retinas were
isolated and stored in pairs at 80°C in preservative (RNAlater; Am-
bion, Inc., Austin, TX). Total RNA was then prepared (RNeasy Minikit;
Qiagen, Inc., Valencia, CA). After quantification at OD
260
, total RNA
was analyzed (RNA Nano LabChips; 2100 Bioanalyzer; Agilent Tech-
nologies, Palo Alto, CA), to assess RNA quality. Only samples showing
minimal degradation were used. cDNA was synthesized (TaqMan Re-
verse Transcription Reagents Kit; ABI) according to the manufacturer’s
instructions. Primers and probes for
-actin and RAGE were designed
on computer (Primer Express; ABI). To confirm specific amplification
of the target mRNA, an aliquot of the PCR product was analyzed by gel
electrophoresis. The sequences of the primers and probe were as
follows: for
-actin, 5-ACG GCC AGG TCA TCA CTA TTG-3 (forward),
5-TGG ATG CCA CAG GAT TCC AT-3 (reverse), and 5-6FAM-ACG
TCT ACC AGC GAA GCT ACT GCC GTC-TAMRA-3 (probe); and for
RAGE, 5-GGA CCC TTA GCT GGC ACT TAG A-3 (forward), 5-GAG
TCC CGT CTC AGG GTG TCT-3 (reverse), and 5-6FAM-ATT CCC GAT
GGC AAA GAA ACA CTC GTG-TAMRA-3 (probe) (ABI). Real-time PCR
was conducted on a sequence-detection system (Prism 7900HTl ABI),
and results were analyzed by the 2
⫺⌬⌬C
T
method.
39
Experiments were
repeated three times, and statistical analysis was performed as de-
scribed later.
Administration of Soluble RAGE
Soluble (s)RAGE, the extracellular two-thirds of the receptor, binds
AGEs and interferes with their ability to bind and activate cellular
RAGE. Preparation, characterization, and purification of sRAGE was
performed with a baculovirus expression system using Sf9 cells (BD-
Clontech, Palo Alto, CA; Invitrogen Corp.) as previously described.
36
Purified murine sRAGE (a single-band of 40 kDa, by Coomassie-
stained SDS-PAGE) was dialyzed against PBS; made free of detectable
endotoxin, based on the Limulus amebocyte assay (E-Toxate; Sigma)
after passage onto gel columns (Detoxi-Gel; Pierce Chemical Co.,
Rockford, IL); and sterile-filtered (0.2
m). We administered daily
doses of 100
g sRAGE based on previous dose–response studies.
27
FIGURE 1. Retinal elastase digest results among diabetic, hyperlipid-
emic, and littermate control mice at age 6 months. The development of
acellular capillaries (A, arrow; B) was accelerated in the retinas of
HGHL mice, with significantly more acellular capillaries present per
unit area than in NGNL or NGHL and HGNL mice. Pericyte ghosts (C,
arrow; D) were also increased in the retinas of HGHL mice compared
with NGNL littermates at age 6 months. Capillary outpouching (E,
arrow), suggesting early microaneurysm formation, was observed in
the retinal vasculature of HGHL mice. An intercapillary bridge, a
normal feature of retinal digests not included in analysis, was also
visible (arrowhead). Results are expressed as the mean SEM. *P
0.05; **P 0.01. Scale bar, 50
m.
TABLE 1. Glucose and Cholesterol Level at Euthanatizing*
NGNL NGHL HGNL HGHL
Glucose (mg/dL) 121.3 28.5 (15) 113.8 24.2 (18) 452.6 109.4 (13) 455.5 68.4 (10)
Cholesterol (mg/dL) 62.5 14.4 (5) 471.2 72.4 (15) 201.3 30.4 (5) 955.6 149.1 (15)
Data are expressed as the mean SD (n).
* Mice were euthanatized at six months.
2918 Barile et al. IOVS, August 2005, Vol. 46, No. 8
Statistical Analysis
To analyze the vascular, neuronal, and experimental data among the
four groups, we used a two-factor analysis of variance (ANOVA) model.
The two factors considered were glucose (normal/high) and lipid
(normal/high). Interactions were tested for all analyses, but none was
found. A one-way ANOVA was also used to compare the four groups in
analyzing the AGE ELISA and autofluorescence data and the RAGE
q-PCR data. For the experiment involving treatment with sRAGE, a
one-way ANOVA was used to compare the three groups: NGNL, HGHL,
and sRAGE. If a difference was found among the groups (P 0.05), a
post hoc analysis using the Duncan test was performed. All data were
analyzed on computer (SAS system software; SAS Institute Inc., Cary,
NC).
RESULTS
Effect of Hyperlipidemia on the Development of
Vascular Lesions of Early Diabetic Retinopathy in
Hyperglycemic Mice
The serum levels of glucose and cholesterol for each of the four
groups are presented in Table 1. We first examined the impact
of the introduction of hyperlipidemia into the hyperglycemic
db/db background on vascular properties in the retina. At age
6 months, the retinas of HGHL (apoE
/
db/db) mice dis
-
played the most significant capillary lesions of NPDR (Fig. 1).
Whereas the eyes of HGNL mice exhibited some development
of acellular capillaries within the retina, only the eyes of HGHL
mice had a significantly higher number of acellular capillaries
than all other groups (Fig. 1B). The development of pericyte
ghosts was detectable in both the HGNL and NGHL pheno-
types, but only in the HGHL mice was there a significant
difference from the NGNL control animals (Fig. 1D). Only in
HGHL mice was there evidence of capillary outpouching con-
sistent with early microaneurysm formation (Fig. 1E).
Electrophysiologic Neural Dysfunction of the
Inner Retina of Hyperglycemic Mice
Electrophysiologic testing at age 6 months revealed that hyper-
glycemia resulted in early inner retinal dysfunction of the
retina detected by prolongation in the latencies of the b-wave
and the OPs (Table 2). Specifically, there were significant
hyperglycemia-induced delays in the implicit time of the b-
wave and OP1, OP2, and OP3 (see Table 4). The ERG ampli-
tudes were not significantly affected in this study, with hyper-
glycemic mice demonstrating a statistically significant decline
only in the amplitude of OP1 (Tables 3, 4). Hyperlipidemia
alone did not induce statistically significant differences in any
of the parameters recorded and studied (Table 4).
The RAGE Axis at the Vitreoretinal Interface
RAGE expression was predominantly localized to glial cells of
the inner retina. Most of the RAGE-expressing cells within the
neural retina were consistent with the distribution of Mu¨ller
cells—particularly their internal footplates. In merged images,
RAGE-positive cells of the inner retina colocalized with vimen-
tin expression, confirming Mu¨ller cell expression (Fig. 2).
GFAP expression in astrocytes of the inner retina revealed no
evidence of colocalization with adjacent RAGE expression of
Mu¨ller cell processes and footplates (Figs. 3A–C). Expression of
RAGE was also detected adjacent to the microvasculature,
suggesting intimate neurovascular localization for RAGE in the
circulation of the inner retina (Figs. 3D, 3E). AGEs were prom-
inently detected within the vitreous cavity of the eye and
particularly along the vitreoretinal interface including the in-
ternal limiting membrane (Figs. 4B, 4F). AGEs were consis-
tently detected within the lens capsule and Bruch’s membrane
and occasionally within the basement membrane of the micro-
vasculature (not shown). In AGE and RAGE merged images,
AGE was localized to vitreous fibrils and the internal limiting
membrane, where there was close apposition to the footplates
of RAGE-expressing Mu¨ller cells (Fig. 4).
RAGE and Its AGE Ligands in NPDR
We next quantified the RAGE axis in this murine model of
NPDR. As AGEs can accumulate within cellular protein as well
as within the proteins of ECM, we assayed the autofluores-
cence of AGEs independently. As shown in Table 5, there was
not a significant difference among groups with regard to AGE
autofluorescence in the cellular protein. In contrast, AGE
autofluorescence increased in ECM in the setting of hypergly-
cemia, but only the retinas of HGHL mice had a significant
difference in fluorescence when compared with NGNL mice.
To quantify AGEs further in the ECM, we performed a non-
competitive ELISA. This study revealed significantly increased
AGE formation in the retinal ECM of hyperglycemic mice, both
HGNL and HGHL (Fig. 5A). As RAGE expression may be am-
plified in the setting of its ligands,
40
RAGE mRNA expression
from whole retina was then examined by quantitative real-time
PCR for each group. RAGE mRNA expression was increased in
TABLE 2. ERG Latencies in Mice at Age 6 Months
NGNL (n 18) NGHL (n 10) HGNL (n 8) HGHL (n 14)
b-Wave 32.0 2.0 32.4 4.0 35.3 3.5 34.5 2.9
OP1 23.4 1.4 23.0 2.1 25.4 1.9 24.6 1.7
OP2 32.0 2.0 31.7 3.2 34.8 2.5 33.8 2.2
OP3 42.6 2.9 42.9 5.4 45.4 3.2 44.9 3.1
OPs 98.0 6.2 97.5 10.6 105.6 7.3 103.3 6.8
Data are expressed as the mean latency SD (ms).
TABLE 3. ERG Amplitudes in Mice at Age 6 Months
NGNL (n 18) NGHL (n 10) HGNL (n 8) HGHL (n 14)
b-Wave 568.4 163.2 517.0 141.0 462.5 138.9 489.3 186.5
OP1 234.6 62.9 210.5 84.4 173.5 52.6 175.6 67.2
OP2 244.5 89.8 206.8 96.6 219.4 54.7 202.9 68.2
OP3 96.2 50.6 80.0 39.8 109.9 32.6 96.0 50.0
OPs 575.3 191.8 497.3 212.1 502.8 109.7 474.5 168.2
Data are expressed as the mean amplitude SD (mV)
IOVS, August 2005, Vol. 46, No. 8 RAGE in Early Diabetic Retinopathy 2919
the retinas of hyperglycemic mice (glucose effect for two-
factor ANOVA: P 0.01); a significant increase was observed
in HGHL mice compared with each group of normoglycemic
mice (Fig. 5B). These studies demonstrate that the RAGE axis
comprising the cellular receptor and its AGE ligands is ampli-
fied in the diabetic retina, particularly in eyes with significant
capillary lesions of NPDR (HGHL mice).
Effect of Antagonism of RAGE on the Vascular
Lesions of Diabetic Retinopathy and Neuronal
Dysfunction at 6 Months of Age
Based on the upregulation of AGEs and RAGE in the HGHL
group, we tested the potential contribution of RAGE to the
pathogenesis of vascular and neuronal perturbation. Murine
sRAGE was administered to 10 HGHL mice from age 8 weeks to
age 6 months. The number of acellular capillaries per 10 mm
2
in the retinal digest of treated mice was significantly less than
those observed in nontreated mice (Fig. 6A). In addition, there
were significantly fewer pericyte ghosts in the retinas of
treated mice than in those of nontreated mice (Fig. 6B). Elec-
trophysiologic studies demonstrated that prophylactic treat-
ment with sRAGE reduced retinal neuronal dysfunction, with a
statistically significant (P 0.05) reduction in the hyperglyce-
mia-induced latency delays observed in OP2, OP3, and OPs
(summation of OPs) at 6 months of age (Table 6). Treatment
with sRAGE had no significant effect on the amplitudes of the
b-wave and OPs (data not shown).
DISCUSSION
The pathogenesis of diabetic retinopathy remains complex,
but prolonged hyperglycemia is essential in the development
of anatomic retinal vascular lesions in human diabetic retinop-
athy and most animal models of diabetic retinopathy.
41
In this
context, we investigated the db/db mouse, a well-character-
ized murine model of hereditary, insulin-resistant diabetes first
detected in the progeny of the C57BLKS/J strain at the Jackson
Laboratory and later characterized as being deficient in leptin
receptor signaling.
42
While the db/db mouse develops neurop
-
athy and nephropathy, the anatomic retinal vascular findings,
apart from basement membrane thickening, are less dramatic.
Our previous anatomic studies revealed acellular capillaries
and pericyte ghosts at age 8 months in db/db mice, but these
anatomic findings were variable and inconsistently present
(Barile GR, et al. IOVS 2000;41:ARVO Abstract 2156). Hyper-
lipidemia is associated with the severity of diabetic retinopa-
thy,
8–10
and successful treatment of hyperlipidemia in diabetic
patients may retard the progression of retinopathy or improve
it.
11–13
For these reasons, we investigated the influence of
hyperlipidemia on the retinal findings of the db/db mouse
model of diabetes mellitus, ultimately crossing it with mice
carrying a mutation in the apoE gene that leaves them devoid
of functioning apoE protein. We observed that the classic
anatomic retinal lesions of nonproliferative diabetic retinopa-
thy developed at the highest rate in HGHL mice, when com-
pared with the other groups, consistent with the burgeoning
notion that hyperlipidemia accelerates the retinal vascular dis-
ease of diabetes mellitus. These results further support increas-
ing evidence that dyslipidemia in diabetes mellitus indepen-
dently contributes to the pathogenesis and severity of diabetic
retinopathy, possibly through amplification of inflammatory
mechanisms.
43,44
Whereas diabetic retinopathy is classically a microvascular
disease of the retinal capillaries, diabetes may impair retinal
neuronal function before the onset of visible vascular lesions.
Numerous psychophysical and electrophysiological studies
demonstrate early retinal neuronal dysfunction in diabetes mel-
litus, before the onset of the classic microvascular lesions of
diabetic retinopathy.
45,46
In particular, Bresnick and Palta
47
and Bresnick
48
have emphasized that alterations in the OPs of
the electroretinogram better predict the development of high-
risk proliferative retinopathy than do clinical fundus photo-
graphs. Pathologic quantification of neural loss by Barber et
al.
49
showed apoptosis of retinal neurons and retinal atrophy,
with loss of inner retinal thickness and cell bodies, in both
diabetic rats and humans. Several investigators have noted
other retinal neuronal alterations in early diabetes, including
GFAP activation and glutamate transporter dysfunction in Mu¨l-
ler cells.
50,51
In our study, chronic hyperglycemia caused sig
-
nificant implicit time delays of OPs at 6 months that are com-
parable to those in previous studies of diabetes,
52
whereas
hyperlipidemia did not influence these electrophysiologic pa-
rameters. In conjunction with the histopathologic vascular
changes observed, this study supports the concept of early
diabetic retinopathy as a neurovascular disease of the retina,
with physiologic disturbances of neuronal function accompa-
nying traditional microvascular capillary pathologic disease.
It was in these contexts that we examined the RAGE axis in
this newly characterized murine model of NPDR. Not surpris-
ingly, we observed prominent AGE localization within the
vitreous cavity. The increased AGE formation in the vitreous
cavity of diabetic eyes has been postulated to increase collagen
cross-linking and cause vitreous changes characteristic of dia-
betic eyes, well-recognized phenomena sometimes referred to
as diabetic vitreoschisis or vitreopathy.
19,20
An additional find
-
ing of our study was prominent AGE accumulation along the
vitreoretinal interface, specifically posterior vitreous cortex
and the internal limiting membrane. Similar to the vitreous
cavity, the accumulation of AGEs at the vitreoretinal interface
may result in structural alterations that promote mechanical
traction in this region. Vitrectomy procedures are sometimes
performed to remove tractional effects that promote diabetic
macular edema. The localization of AGEs along the vitreoreti-
nal interface is consistent with the concept of a structurally
altered posterior hyaloid and internal limiting membrane capa-
ble of promoting subclinical vitreomacular disease in early
diabetic retinopathy. AGEs may also exert nontractional, recep-
tor-mediated effects through the RAGE axis. In this regard, an
intriguing finding of our study is the localization of RAGE
primarily to the Mu¨ller cells that extend from the internal
limiting membrane to the external limiting membrane of the
retina. The anatomically close apposition of an AGE-laden in-
ternal limiting membrane with the RAGE-expressing footplates
suggests that a possible physiologic benefit of diabetic vitrec-
tomy is the removal of AGE ligands from the posterior vitreous
TABLE 4. Two-Factor ANOVA Analysis of ERG Data from Tables 2
and 3
Glucose Effect Lipid Effect Interaction
b-Wave
Latency 0.004 0.805 0.516
Amplitude 0.174 0.799 0.422
OP1
Latency 0.001 0.216 0.649
Amplitude 0.021 0.588 0.516
OP2
Latency 0.001 0.376 0.675
Amplitude 0.550 0.225 0.661
OP3
Latency 0.031 0.933 0.744
Amplitude 0.283 0.271 0.934
OPs
Latency 0.004 0.545 0.685
Amplitude 0.376 0.324 0.643
Data are probabilities.
2920 Barile et al. IOVS, August 2005, Vol. 46, No. 8
FIGURE 2. RAGE expression in the
retina of NGNL and HGHL mice.
RAGE immunofluorescence (A, D,
green) colocalizes with vimentin (B,
E, red), a marker of Mu¨ller cells (ar-
rows) in both NLNG and HGHL mice
(C, F). The extension of Mu¨ller cells
from the internal to the external lim-
iting membranes of the neurosensory
was highlighted with RAGE’s expres-
sion (A, D). ILM, internal limiting
membrane; IPL, inner plexiform lay-
er; INL, inner nuclear layer; ONL,
outer nuclear layer; ELM, external
limiting membrane. Scale bar, 50
m.
FIGURE 3. RAGE, GFAP, and CD31
immunohistochemistry of the retina
of HGHL mice. RAGE expression was
prominent in Mu¨ller cell processes,
particularly their internal footplates
(A, D; green, arrowheads) and was
not observed in adjacent astrocytes
(B, C; red, arrows). The intimate va-
soglial relationship of the RAGE-ex-
pressing Mu¨ller cell (D, green) with
the vascular endothelium of a retinal
capillary (E, red) is observed in (F).
ILM, internal limiting membrane; IPL,
inner plexiform layer; INL, inner nu-
clear layer. Scale bar, 25
m.
FIGURE 4. RAGE (green) and AGE
(red) immunohistochemistry of the
vitreoretinal interface in NGNL mice
(AC) and HGHL mice (EG). AGEs
are detected within the vitreous cav-
ity, posterior vitreous cortex, and in-
ternal limiting membrane of the ret-
ina (B, F, red). The internal
footplates of RAGE-expressing Mu¨ller
cells (A, E, green) are immediately
adjacent to AGEs in the internal lim-
iting membrane (C, G). Controls (D,
H). Vit, vitreous cavity; ILM, internal
limiting membrane; GCL, ganglion
cell layer; IPL, inner plexiform layer;
INL, inner nuclear layer. Scale bar, 25
m.
IOVS, August 2005, Vol. 46, No. 8 RAGE in Early Diabetic Retinopathy 2921
cortex and internal limiting membrane, downregulating the
proinflammatory RAGE axis in adjacent Mu¨ller cells.
The localization of RAGE to Mu¨ller cells raises exciting
possibilities for novel roles of these cells in the pathogenesis of
diabetic retinopathy. The specific RAGE-dependent mecha-
nisms by which AGEs may alter Mu¨ller cell structure and
function are the subject of future study. These cells are well
known to display a varied repertoire of structural and physio-
logic properties in the retina. The contact of vasoglial neuronal
tissue and especially Mu¨ller cells with underlying capillaries in
the retina suggests a potential pathophysiologic relationship in
diabetic retinopathy, once suggested by Ashton in his Bowman
lecture and supported by several recent studies.
53
In the set
-
ting of diabetes, alteration of the glutamate transporter, in part,
it is speculated, by oxidation; increased expression of GFAP
suggestive of reactive gliosis; and striking upregulation of VEGF
all have been detected in Mu¨ller cells.
54
Indeed, in vitro anal
-
yses suggests that incubation of cultured Mu¨ller cells with
AGEs upregulate expression of VEGF.
55
Mu¨ller cell ischemia
induces phosphorylation of extracellular signal-regulated ki-
nase (ERK) MAPKs in these cells,
56
again suggesting that a wide
array of changes in gene expression may ensue in these cells
when perturbed. The possible RAGE-dependence of these phe-
nomena remains to be determined, but the intimate relation-
ship of RAGE-expressing Mu¨ller cells with underlying vascular
endothelium suggests a potential role for Mu¨ller cell RAGE in
neurovascular dysfunction.
In addition to the specific localization of RAGE and its AGE
ligands in our study, we observed that AGEs accumulate in the
neurosensory retina with associated amplification of cellular
RAGE in the setting of hyperglycemia and early diabetic reti-
nopathy. The diversity by which AGEs may form on the amino
groups of proteins, lipids, and DNA is reflected in the variety of
locations that these products may accumulate during hypergly-
cemia, including the serum, ECM, and intracellular cyto-
plasm.
19
In this regard, it is noteworthy that we did not detect
significantly different AGE levels by fluorescence studies
within cellular proteins among the hyperlipidemic and hyper-
glycemic phenotypes. Instead, the retinas with the most severe
capillary disease had the highest levels of AGEs detected within
the ECM, both by fluorescence and ELISA studies. Hyperglyce-
FIGURE 5. Retinal AGE ELISA and RAGE mRNA transcripts. Retinal
AGEs accumulated in the retinas of hyperglycemic mice: Both HGNL
mice and HGHL mice had significantly increased AGEs compared with
NGNL littermates (A). RAGE mRNA expression in the retina was
increased in the setting of hyperglycemia and AGE accumulation.
RAGE transcripts were highest in the retinas of HGHL mice, with a
nearly twofold elevation compared with basal levels in NGNL litter-
mates as well as a significant increase compared with NGHL mice (B).
Results are expressed as the mean SEM. *P 0.05; **P 0.01.
FIGURE 6. Effect of RAGE antagonism on vascular changes in HGHL
mice. sRAGE-treated mice had significantly less acellular capillaries (A)
and pericyte ghosts (B) in the retina compared with untreated HGHL
mice. Treatment of these mice also reduced the latency delays ob-
served in the OPs, with a significant reduction in the implicit times of
OP2, OP3 and OPs (the summation of OPs). *P 0.05.
TABLE 5. Retinal AGE Fluorescence Intensities
NGNL NGHL HGNL HGHL
Cellular protein 1357 149 1666 182 1122 194 1181 161
ECM 2801 673 2342 531 3713 1229 5259 715*
Data are expressed as the mean autofluorescence SE.
* Significant difference (P 0.05) compared to the NGNL group.
2922 Barile et al. IOVS, August 2005, Vol. 46, No. 8
mia was the most important contributor to the development of
these AGEs, as HGNL mice also exhibited increased AGE accu-
mulation in the ECM in these studies (though this increase was
shown to be significant in this group only by ELISA). Consistent
with a role for RAGE ligands such as AGEs in the development
of retinopathy, we detected significant upregulation of RAGE
transcripts in the retinas of HGHL mice that had the highest
AGE accumulation and retinal disease. The amplification of
RAGE in the setting of its ligands is consistent with the known
biology of RAGE in other organ systems, and this property mag-
nifies the effect of the RAGE axis in local cellular responses.
26,40
Importantly, in this study, antagonism of the RAGE axis
ameliorated both neuronal dysfunction and vascular disease.
The electrophysiologic benefit we observed suggests that
RAGE contributes to neuronal dysfunction in the diabetic ret-
ina. The mechanisms of OP generation in the normal retina, the
associated alterations observed in these neuronal responses in
diabetic eyes, and the extent to which altered Mu¨ller cell
glutamate metabolism, signaling, and gene expression contrib-
ute to perturbation of these signals remains to be determined.
Antagonism of RAGE also reduced the progression of vascular
lesions of diabetic retinopathy in HGHL mice. This vascular
effect may relate to an a priori neuronal benefit to RAGE-
expressing Mu¨ller cells, but the ample data on AGE toxicity and
perturbation in retinal vascular endothelial cells also suggests
that antagonism of circulating serum AGEs with sRAGE may
reduce these perturbations and the resultant anatomic disease.
The precise neurovascular mechanisms altered with ligand
interaction with RAGE in the retina remain the subject of
current and future investigations, but the amelioration of neu-
rovascular features of diabetic retinopathy observed in this
study identifies the RAGE axis as an important therapeutic
target in the prevention and treatment of diabetic complica-
tions in the retina.
References
1. Frank RN. Diabetic retinopathy. N Engl J Med. 2004;350:48 –58.
2. Gardner TW, Antonetti DA, Barber AJ, LaNoue KF, Levison SW.
Diabetic retinopathy: more than meets the eye. Surv Ophthalmol.
2002;47(suppl 2):S253–S262.
3. Lieth E, Gardner TW, Barber AJ, Antonetti DA. Retinal
neurodegeneration: early pathology in diabetes. Clin Exp Ophthal-
mol. 2000;28:3–8.
4. Aiello LP, Avery RL, Arrigg PG, et al. Vascular endothelial growth
factor in ocular fluid of patients with diabetic retinopathy and
other retinal disorders. N Engl J Med. 1994;331:1480 –1487.
5. The Diabetes Control and Complications Trial Research Group.
The effect of intensive treatment of diabetes on the development
and progression of long-term complications in insulin-dependent
diabetes mellitus. N Engl J Med. 1993;329:977–986.
6. The UK Prospective Diabetes Study (UKPDS) Group. Intensive
blood-glucose control with sulphonylureas or insulin compared
with conventional treatment and risk of complications in patients
with type 2 diabetes (UKPDS 33). Lancet. 1998;352:837– 853.
7. The UK Prospective Diabetes Study (UKPDS) Group. Tight blood
pressure control and risk of macrovascular and microvascular
complications in type 2 diabetes: UKPDS 38. BMJ. 1998;317:703–
713.
8. Kissebah AH, Kohner EM, Lewis B, Siddiq YK, Lowy C, Fraser TR.
Plasma-lipids and glucose/insulin relationship in non-insulin-requir-
ing diabetics with and without retinopathy. Lancet. 1975;1:1104
1108.
9. Chew EY, Klein ML, Ferris FL III, et al. Association of elevated
serum lipid levels with retinal hard exudate in diabetic retinopa-
thy. Early Treatment Diabetic Retinopathy Study (ETDRS) Report
22. Arch Ophthalmol. 1996;114:1079 –1084.
10. Davis MD, Fisher MR, Gangnon RE, et al. Risk factors for high-risk
proliferative diabetic retinopathy and severe visual loss: Early
Treatment Diabetic Retinopathy Study Report #18. Invest Oph-
thalmol Vis Sci. 1998;39:233–252.
11. Sen K, Misra A, Kumar A, Pandey RM. Simvastatin retards progres-
sion of retinopathy in diabetic patients with hypercholesterolemia.
Diabetes Res Clin Pract. 2002;56:1–11.
12. Gordon B, Chang S, Kavanagh M, et al. The effects of lipid lower-
ing on diabetic retinopathy. Am J Ophthalmol. 1991;112:385–391.
13. Fried LF, Forrest KY, Ellis D, Chang Y, Silvers N, Orchard TJ. Lipid
modulation in insulin-dependent diabetes mellitus: effect on mi-
crovascular outcomes. J Diabetes Complications. 2001;15:113–
119.
14. Schleicher ED, Wagner E, Nerlich AG. Increased accumulation of
the glycoxidation product N(epsilon)-(carboxymethyl)lysine in hu-
man tissues in diabetes and aging. J Clin Invest. 1997;99:457– 468.
15. Hammes HP, Alt A, Niwa T, et al. Differential accumulation of
advanced glycation end products in the course of diabetic retinop-
athy. Diabetologia. 1999;42:728 –736.
16. Lu M, Kuroki M, Amano S, et al. Advanced glycation end products
increase retinal vascular endothelial growth factor expression.
J Clin Invest. 1998;101:1219 –1224.
17. Tolentino MJ, Miller JW, Gragoudas ES, et al. Intravitreous injec-
tions of vascular endothelial growth factor produce retinal isch-
emia and microangiopathy in an adult primate. Ophthalmology.
1996;103:1820–1828.
18. Moore TC, Moore JE, Kaji Y, et al. The role of advanced glycation
end products in retinal microvascular leukostasis. Invest Ophthal-
mol Vis Sci. 2003;44:4457– 4464.
19. Stitt AW. The role of advanced glycation in the pathogenesis of
diabetic retinopathy. Exp Mol Pathol. 2003;75:95–108.
20. Sebag J. Abnormalities of human vitreous structure in diabetes.
Graefes Arch Clin Exp Ophthalmol. 1993;231:257–260.
21. Kern TS, Engerman RL. Pharmacological inhibition of diabetic
retinopathy: aminoguanidine and aspirin. Diabetes. 2001;50:
1636–1642.
22. Gardiner TA, Anderson HR, Stitt AW. Inhibition of advanced gly-
cation end-products protects against retinal capillary basement
membrane expansion during long-term diabetes. J Pathol. 2003;
201:328–333.
23. Stitt A, Gardiner TA, Alderson NL, et al. The AGE inhibitor pyri-
doxamine inhibits development of retinopathy in experimental
diabetes. Diabetes. 2002;51:2826 –2832.
24. Hammes HP, Du X, Edelstein D, et al. Benfotiamine blocks three
major pathways of hyperglycemic damage and prevents experi-
mental diabetic retinopathy. Nat Med. 2003;9:294–299.
25. Schmidt AM, Vianna M, Gerlach M, et al. Isolation and character-
ization of two binding proteins for advanced glycosylation end
products from bovine lung which are present on the endothelial
cell surface. J Biol Chem. 1992;267:14987–14997.
26. Yan SF, Ramasamy R, Naka Y, Schmidt AM. Glycation, inflamma-
tion, and RAGE: a scaffold for the macrovascular complications of
diabetes and beyond. Circ Res. 2003;93:1159–1169.
27. Hofmann MA, Drury S, Fu C, et al. RAGE mediates a novel proin-
flammatory axis: A central cell surface receptor for s100/calgranu-
lin polypeptides. Cell. 1999;97:889 –901.
28. Hori O, Brett J, Slattery T, et al. The receptor for advanced glyca-
tion end products (RAGE) is a cellular binding site for amphoterin:
mediation of neurite outgrowth and co-expression of RAGE and
amphoterin in the developing nervous system. J Biol Chem. 1995;
270:25752–25761.
29. Chavakis T, Bierhaus A, Al-Fakhri N, et al. The pattern recognition
receptor (RAGE) is a counterreceptor for leukocyte integrins: a
novel pathway for inflammatory cell recruitment. J Exp Med.
2003;198:1507–1515.
T
ABLE 6. sRAGE Effect on ERG Latencies at Age 6 Months
NGNL (n 18) HGHL (n 14) sRAGE (n 10)
b-Wave 32.0 2.0 34.5 2.9 33.3 2.5
OP1 23.4 1.4 24.6 1.7 23.6 1.4
OP2 32.0 2.0 33.8 2.2 32.2 2.0*
OP3 42.6 2.9 44.9 3.1 42.0 3.0*
OPs 98.0 6.2 103.3 6.8 97.7 6.1*
Data are expressed as the mean latency SD (ms).
* Significant (P 0.05) compared to HGHL group.
IOVS, August 2005, Vol. 46, No. 8 RAGE in Early Diabetic Retinopathy 2923
30. Schmidt AM, Hori O, Chen JX, et al. Advanced glycation end
products interacting with their endothelial receptor induce ex-
pression of vascular cell adhesion molecule-1 (VCAM-1) in cul-
tured human endothelial cells and in mice: a potential mechanism
for the accelerated vasculopathy of diabetes. J Clin Invest. 1995;
96:1395–1403.
31. Wautier JL, Zoukourian C, Chappey O, et al. Receptor-mediated
endothelial cell dysfunction in diabetic vasculopathy: soluble re-
ceptor for advanced glycation end products blocks hyperperme-
ability in diabetic rats. J Clin Invest. 1996;97:238 –243.
32. Laver NM, Robison WG Jr, Pfeffer BA. Novel procedures for iso-
lating intact retinal vascular beds from diabetic humans and animal
models. Invest Ophthalmol Vis Sci. 1993;34:2097–2104.
33. Kuwabara T, Cogan DG. Studies of retinal vascular patterns. I.
Normal architecture. Arch Ophthalmol. 1960;64:904 –911.
34. Algvere P. Clinical studies on the oscillatory potentials of the
human electroretinogram with special reference to the scotopic
b-wave. Acta Ophthalmol (Copenh). 1968;46:993–1024.
35. Segawa M, Hirata Y, Fujimori S, Okada K. The development of
electroretinogram abnormalities and the possible role of polyol
pathway activity in diabetic hyperglycemia and galactosemia. Me-
tabolism. 1988;37:454460.
36. Park L, Raman KG, Lee KJ, et al. Suppression of accelerated
diabetic atherosclerosis by the soluble receptor for advanced gly-
cation endproducts. Nat Med. 1998;4:1025–1031.
37. Yin D. Biochemical basis of lipofuscin, ceroid, and age pigment-
like fluorophores. Free Radic Biol Med. 1996;21:871–888.
38. Meerwaldt R, Graaff R, Oomen PH, et al. Simple non-invasive
assessment of advanced glycation endproduct accumulation. Dia-
betologia. 2004;47:1324–1330.
39. Livak KJ, Schmittgen TD. Analysis of relative gene expression data
using real-time quantitative PCR and the 2(-Delta Ddelta C(T))
method. Methods. 2001;25:402– 408.
40. Li J, Schmidt AM. Characterization and functional analysis of the
promoter of RAGE, the receptor for advanced glycation end prod-
ucts. J Biol Chem. 1997;272:16498 –16506.
41. Engerman RL, Kern TS. Retinopathy in animal models of diabetes.
Diabetes Metab Rev. 1995;11:109 –120.
42. Chua SC Jr, Chung WK, Wu-Peng XS, et al. Phenotypes of mouse
diabetes and rat fatty due to mutations in the OB (leptin) receptor.
Science. 1996;271:994–996.
43. Lyons TJ, Jenkins AJ, Zheng D, et al. Diabetic retinopathy and
serum lipoprotein subclasses in the DCCT/EDIC cohort. Invest
Ophthalmol Vis Sci. 2004;45:910 –918.
44. Chen W, Jump DB, Grant MB, Esselman WJ, Busik JV. Dyslipide-
mia, but not hyperglycemia, induces inflammatory adhesion mol-
ecules in human retinal vascular endothelial cells. Invest Ophthal-
mol Vis Sci. 2003;44:5016 –5022.
45. Tzekov R, Arden GB. The electroretinogram in diabetic retinopa-
thy. Surv Ophthalmol. 1999;44:53– 60.
46. Han Y, Bearse MA Jr, Schneck ME, Barez S, Jacobsen CH, Adams AJ.
Multifocal electroretinogram delays predict sites of subsequent
diabetic retinopathy. Invest Ophthalmol Vis Sci. 2004;45:948
954.
47. Bresnick GH, Palta M. Predicting progression to severe prolifera-
tive diabetic retinopathy. Arch Ophthalmol. 1987;105:810 814.
48. Bresnick GH. Diabetic retinopathy viewed as a neurosensory dis-
order. Arch Ophthalmol. 1986;104:989 –990.
49. Barber AJ, Lieth E, Khin SA, Antonetti DA, Buchanan AG, Gardner
TW. Neural apoptosis in the retina during experimental and hu-
man diabetes. Early onset and effect of insulin. J Clin Invest.
1998;102:783–791.
50. Zeng XX, Ng YK, Ling EA. Neuronal and microglial response in the
retina of streptozotocin-induced diabetic rats. Vis Neurosci. 2000;
17:463–471.
51. Mizutani M, Gerhardinger C, Lorenzi M. Mu¨ller cell changes in
human diabetic retinopathy. Diabetes. 1998;47:445– 449.
52. Shirao Y, Kawasaki K. Electrical responses from diabetic retina.
Prog Retin Eye Res. 1998;17:59 –76.
53. Ashton N. Bowman lecture: the blood-retinal barrier and vaso-glial
relationships in retinal disease. Trans Ophthalmol Soc UK. 1965;
85:199–230.
54. Amin RH, Frank RN, Kennedy A, Eliott D, Puklin JE, Abrams GW.
Vascular endothelial growth factor is present in glial cells of the
retina and optic nerve of human subjects with nonproliferative
diabetic retinopathy. Invest Ophthalmol Vis Sci. 1997;38:36 47.
55. Hirata C, Nakano K, Nakamura N, et al. Advanced glycation end
products induce expression of vascular endothelial growth factor
by retinal Mu¨ller cells. Biochem Biophys Res Commun. 1997;236:
712–715.
56. Roth S, Shaikh AR, Hennelly MM, Li Q, Bindokas V, Graham CE.
Mitogen-activated protein kinases and retinal ischemia. Invest
Ophthalmol Vis Sci. 2003;44:5383–5395.
2924 Barile et al. IOVS, August 2005, Vol. 46, No. 8
... Inhibition in AGE formation through administration of aminoguanidine or pyridoxamine inhibits retinal pathologies in diabetic rodents [17,22]. In addition, attenuation of the effects of AGEs through administration of soluble receptors for AGEs reduces blood-retinal barrier breakdown, leukostasis, the expression of ICAM-1, neuronal dysfunction, and the development of capillary lesions in diabetic mice [23,24]. ...
... AGEs have been linked to various inflammatory responses that are key to the development of diabetic retinopathy. These include ICAM-1 upregulation, leukostasis, vascular leakage, apoptosis, and the development of capillary lesions [20,21,23,24,39]. Interestingly, CD40 promotes ICAM-1 upregulation, leukostasis, and retinal capillary degeneration, and is required for the development of diabetic retinopathy [2]. ...
Article
Full-text available
CD40 induces pro-inflammatory responses in endothelial and Müller cells and is required for the development of diabetic retinopathy (DR). CD40 is upregulated in these cells in patients with DR. CD40 upregulation is a central feature of CD40-driven inflammatory disorders. What drives CD40 upregulation in the diabetic retina remains unknown. We examined the role of advanced glycation end products (AGEs) in CD40 upregulation in endothelial cells and Müller cells. Human endothelial cells and Müller cells were incubated with unmodified or methylglyoxal (MGO)-modified fibronectin. CD40 expression was assessed by flow cytometry. The expression of ICAM-1 and CCL2 was examined by flow cytometry or ELISA after stimulation with CD154 (CD40 ligand). The expression of carboxymethyl lysine (CML), fibronectin, and laminin as well as CD40 in endothelial and Müller cells from patients with DR was examined by confocal microscopy. Fibronectin modified by MGO upregulated CD40 in endothelial and Müller cells. CD40 upregulation was functionally relevant. MGO-modified fibronectin enhanced CD154-driven upregulation of ICAM-1 and CCL2 in endothelial and Müller cells. Increased CD40 expression in endothelial and Müller cells from patients with DR was associated with increased CML expression in fibronectin and laminin. These findings identify AGEs as inducers of CD40 upregulation in endothelial and Müller cells and enhancers of CD40-dependent pro-inflammatory responses. CD40 upregulation in these cells is associated with higher CML expression in fibronectin and laminin in patients with DR. This study revealed that CD40 and AGEs, two important drivers of DR, are interconnected.
... Impaired angiogenesis and neovascularization are common features of diabetic vascular complications, and AGEs have been implicated in these processes. AGEs can inhibit the proliferation, migration, and tube formation of endothelial progenitor cells and impair the release of proangiogenic factors, such as vascular endothelial growth factor (VEGF) [36,37]. Furthermore, AGEs can activate the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway, which inhibits endothelial cell proliferation and contributes to impaired angiogenesis [38]. ...
Article
Full-text available
Background In diabetic metabolic disorders, advanced glycation end products (AGEs) contribute significantly to the development of cardiovascular diseases (CVD). Aims This comprehensive review aims to elucidate the molecular mechanisms underlying AGE-mediated vascular injury. Conclusions We discuss the formation and accumulation of AGEs, their interactions with cellular receptors, and the subsequent activation of signaling pathways leading to oxidative stress, inflammation, endothelial dysfunction, smooth muscle cell proliferation, extracellular matrix remodeling, and impaired angiogenesis. Moreover, we explore potential therapeutic strategies targeting AGEs and related pathways for CVD prevention and treatment in diabetic metabolic disorders. Finally, we address current challenges and future directions in the field, emphasizing the importance of understanding the molecular links between AGEs and vascular injury to improve patient outcomes.
... The role of sRAGE is still debated because the available results are contradictory. In animal studies, administration of sRAGE prevented and slowed atherosclerosis [123], improved retinal neuronal dysfunction in diabetic mice [124], and accelerated wound healing in diabetic mice [125,126]. These results suggest that sRAGE acts by trapping, binding, and eliminating circulating AGEs. ...
Article
Full-text available
Ageing is a composite process that involves numerous changes at the cellular, tissue, organ and whole-body levels. These changes result in decreased functioning of the organism and the development of certain conditions, which ultimately lead to an increased risk of death. Advanced glycation end products (AGEs) are a family of compounds with a diverse chemical nature. They are the products of non-enzymatic reactions between reducing sugars and proteins, lipids or nucleic acids and are synthesised in high amounts in both physiological and pathological conditions. Accumulation of these molecules increases the level of damage to tissue/organs structures (immune elements, connective tissue, brain, pancreatic beta cells, nephrons, and muscles), which consequently triggers the development of age-related diseases, such as diabetes mellitus, neurodegeneration, and cardiovascular and kidney disorders. Irrespective of the role of AGEs in the initiation or progression of chronic disorders, a reduction in their levels would certainly provide health benefits. In this review, we provide an overview of the role of AGEs in these areas. Moreover, we provide examples of lifestyle interventions, such as caloric restriction or physical activities, that may modulate AGE formation and accumulation and help to promote healthy ageing.
... GNPs are biocompatible and non-toxic, and their presence can be evaluated with imaging modalities including computed tomography and positron emission tomography [18]. Their use in ophthalmology has hardly been explored. ...
Article
Full-text available
In this study, we characterized diabetic retinopathy in two mouse models and the response to anti-vascular endothelial growth factor (VEGF) injection. The study was conducted in 58 transgenic, non-obese diabetic (NOD) mice with spontaneous type 1 diabetes (n = 30, DMT1-NOD) or chemically induced (n = 28, streptozotocin, STZ-NOD) type 1 diabetes and 20 transgenic db/db mice with type 2 diabetes (DMT2-db/db); 30 NOD and 8 wild-type mice served as controls. Mice were examined at 21 days for vasculopathy, retinal thickness, and expression of genes involved in oxidative stress, angiogenesis, gliosis, and diabetes. The right eye was histologically examined one week after injection of bevacizumab, ranibizumab, saline, or no treatment. Flat mounts revealed microaneurysms and one apparent area of tufts of neovascularization in the diabetic retina. Immunostaining revealed activation of Müller glia and prominent Müller cells. Mean retinal thickness was greater in diabetic mice. RAGE increased and GFAP decreased in DMT1-NOD mice; GFAP and SOX-9 mildly increased in db/db mice. Anti-VEGF treatment led to reduced retinal thickness. Retinas showed vasculopathy and edema in DMT1-NOD and DMT2-db/db mice and activation of Müller glia in DMT1-NOD mice, with some response to anti-VEGF treatment. Given the similarity of diabetic retinopathy in mice and humans, comparisons of type 1 and type 2 diabetic mouse models may assist in the development of new treatment modalities.
... The AGE-specific receptor is the most important signaling receptor, often abbreviated as RAGE. An increase in inflammatory signaling is triggered when AGEs are attached to their receptor, which is known as the RAGE-AGE axis [55]. NF-kB and AP-1 phosphorylation or activation are connected to the AGE-RAGE axis via RAS/MAPK/ERK1/2 signaling pathways. ...
Article
Full-text available
The goal of this review is to increase public knowledge of the etiopathogenesis of diabetic eye diseases (DEDs), such as diabetic retinopathy (DR) and ocular angiosarcoma (ASO), and the likelihood of blindness among elderly widows. A widow’s life in North India, in general, is fraught with peril because of the economic and social isolation it brings, as well as the increased risk of death from heart disease, hypertension, diabetes, depression, and dementia. Neovascularization, neuroinflammation, and edema in the ocular tissue are hallmarks of the ASO, a rare form of malignant tumor. When diabetes, hypertension, and aging all contribute to increased oxidative stress, the DR can proceed to ASO. Microglia in the retina of the optic nerve head are responsible for causing inflammation, discomfort, and neurodegeneration. Those that come into contact with them will get blind as a result of this. Advanced glycation end products (AGE), vascular endothelial growth factor (VEGF), protein kinase C (PKC), poly-ADP-ribose polymerase (PARP), metalloproteinase9 (MMP9), nuclear factor kappaB (NFkB), program death ligand1 (PDL-1), factor VIII (FVIII), and von Willebrand factor (VWF) are potent agents for ocular neovascularisation (ONV), neuroinflammation and edema in the ocular tissue. AGE/VEGF, DAG/PKC, PARP/NFkB, RAS/VEGF, PDL-1/PD-1, VWF/FVIII/VEGF, and RAS/VEGF are all linked to the pathophysiology of DEDs. The interaction between ONV and ASO is mostly determined by the VWF/FVIII/VEGF and PDL-1/PD-1 axis. This study focused on retinoprotective medications that can pass the blood-retinal barrier and cure DEDs, as well as the factors that influence the etiology of neovascularization and neuroinflammation in the eye.
... Hence the retinal local regulation is probably driven by the retinal glia, i.e. Müller cells [52] and thus likely independent of the systemic lipid metabolism [53], the impact of local apolipoprotein levels in the different stages of DR may well be assessed on a local level. ...
Article
Full-text available
An imbalance of plasma apolipoproteins has been linked to diabetic retinopathy (DR); however, there is scarce information regarding their presence in the aqueous humor (AH) and their role in DR. Here, we aimed at analysing the relationship between apolipoprotein concentrations in human AH and the severity of DR. Concentrations of apolipoproteins were measured retrospectively in patients with type 2 diabetes mellitus (T2DM) without DR ( n = 23 ), with mild to moderate nonproliferative DR (NPDR) ( n = 13 ), and advanced NPDR/proliferative DR (PDR) ( n = 14 ) using a multiplex immunoassay. Compared to the non-apparent DR group, the concentrations of seven apolipoproteins were elevated in advanced NPDR/PDR (Apo AI 5.8-fold, Apo AII 4.5-fold, Apo CI 3.3-fold, Apo CIII 6.8-fold, Apo D 3.3-fold, Apo E 2.4-fold, and Apo H 6.6-fold). No significant differences were observed in apolipoprotein concentrations between patients with non-apparent DR and healthy controls ( n = 17 ). In conclusion, the AH concentrations of apolipoproteins AI, AII, CI, CIII, D, E, and H increased in advancing stages of DR, suggesting their role in the pathogenesis of DR, which deserves further examination.
... The AGE-speci c receptor is the most important signaling receptor, often abbreviated as RAGE. An increase in in ammatory signaling is triggered when AGEs are attached to their receptor, which is known as the RAGE-AGE axis [55]. NF-kB and AP-1 phosphorylation or activation are connected to the AGE-RAGE axis via RAS/MAPK/ERK1/2 signaling pathways. ...
Preprint
Full-text available
The goal of this review is to increase public knowledge of the etiopathogenesis of diabetic eye diseases (DEDs), such as diabetic retinopathy (DR) and ocular angiosarcoma (ASO), and the likelihood of blindness among elderly widows. A widow's life in North India, in general, is fraught with peril because of the economic and social isolation it brings, as well as the increased risk of death from heart disease, hypertension, diabetes, depression, and dementia. Neovascularization, neuroinflammation, and edema in the ocular tissue are hallmarks of the ASO, a rare form of malignant tumor. When diabetes, hypertension, and aging all contribute to increased oxidative stress, the DR can proceed to ASO. Microglia in the retina of the optic nerve head are responsible for causing inflammation, discomfort, and neurodegeneration. Those that come into contact with them will get blind as a result of this. Advanced glycation end products ( AGE), vascular endothelial growth factor (VEGF), protein kinase C (PKC), poly-ADP-ribose polymerase (PARP), metalloproteinase9 (MMP9), nuclear factor kappaB (NFkB), program death ligand1 (PDL-1), factor VIII (FVIII), and von Willebrand factor (VWF) are potent agents for neovascularisation (NV), neuroinflammation and edema in the ocular tissue. AGE/VEGF, DAG/PKC, PARP/NFkB, RAS/VEGF, PDL-1/PD-1, VWF/FVIII/VEGF, and RAS/VEGF are all linked to the pathophysiology of DEDs. The interaction between NV and ASO is mostly determined by the VWF/FVIII/VEGF and PDL-1 /PD-1 axis. This study focused on retinoprotective medications that can pass the blood-retinal barrier and cure DEDs, as well as the factors that influence the etiology of neovascularization and neuroinflammation in the eye.
Article
Full-text available
Diabetic retinopathy (DR) is the most common complication that develops in patients with diabetes mellitus (DM) and is the leading cause of blindness worldwide. Fortunately, sight-threatening forms of DR develop only after several decades of DM. This well-documented resilience to DR suggests that the retina is capable of protecting itself from DM-related damage and also that accumulation of such damage occurs only after deterioration of this resilience. Despite the enormous translational significance of this phenomenon, very little is known regarding the nature of resilience to DR. Rodent models of DR have been used extensively to study the nature of the DM-induced damage, i.e., cardinal features of DR. Many of these same animal models can be used to investigate resilience because DR is delayed from the onset of DM by several weeks or months. The purpose of this review is to provide a comprehensive overview of the literature describing the use of rodent models of DR in type-1 and type-2 diabetic animals, which most clearly document the delay between the onset of DM and the appearance of DR. These readily available experimental settings can be used to advance our current understanding of resilience to DR and thereby identify biomarkers and targets for novel, prevention-based approaches to manage patients at risk for developing DR.
Article
Full-text available
Diabetic retinopathy, characterized as a microangiopathy and neurodegenerative disease, is the leading cause of visual impairment in diabetic patients. Many clinical features observed in diabetic retinopathy, such as capillary occlusion, acellular capillaries and retinal non-perfusion, aggregate retinal ischemia and represent relatively late events in diabetic retinopathy. In fact, retinal microvascular injury is an early event in diabetic retinopathy involving multiple biochemical alterations, and is manifested by changes to the retinal neurovascular unit and its cellular components. Currently, intravitreal anti-vascular endothelial growth factor therapy is the first-line treatment for diabetic macular edema, and benefits the patient by decreasing the edema and improving visual acuity. However, a significant proportion of patients respond poorly to anti-vascular endothelial growth factor treatments, indicating that factors other than vascular endothelial growth factor are involved in the pathogenesis of diabetic macular edema. Accumulating evidence confirms that low-grade inflammation plays a critical role in the pathogenesis and development of diabetic retinopathy as multiple inflammatory factors, such as interleukin-1β, monocyte chemotactic protein-1 and tumor necrosis factor -α, are increased in the vitreous and retina of diabetic retinopathy patients. These inflammatory factors, together with growth factors such as vascular endothelial growth factor, contribute to blood-retinal barrier breakdown, vascular damage and neuroinflammation, as well as pathological angiogenesis in diabetic retinopathy, complicated by diabetic macular edema and proliferative diabetic retinopathy. In addition, retinal cell types including microglia, Müller glia, astrocytes, retinal pigment epithelial cells, and others are activated, to secrete inflammatory mediators, aggravating cell apoptosis and subsequent vascular leakage. New therapies, targeting these inflammatory molecules or related signaling pathways, have the potential to inhibit retinal inflammation and prevent diabetic retinopathy progression. Here, we review the relevant literature to date, summarize the inflammatory mechanisms underlying the pathogenesis of diabetic retinopathy, and propose inflammation-based treatments for diabetic retinopathy and diabetic macular edema.
Chapter
The unique structural organization of the retinal microvasculature is essential for the healthy maintenance of the retina. Primarily responsible for the regulation of vascular permeability within this tissue, the blood–retinal barrier (BRB) is a physiological barrier essential for normal visual function. Tightly regulating fluid and electrolyte balance in the surrounding tissue, the BRB is formed by the interaction between specialized cells and the underlying basement membrane. In diabetes, the hyperglycemic environment causes the alteration of this barrier. Unable to regulate vascular permeability, the breakdown of the BRB leads to the leakage of plasma and lipids into the retina, the clinical manifestation of diabetic macular edema (DME). In this chapter, we discuss the key players (VEGF and molecules beyond VEGF) involved in the maintenance of this barrier and the molecular mechanisms that lead to its breakdown. Known to be involved in this disease, we highlight the role of inflammation in DME. Current available therapies have limitations; thus, we point to the various pro-inflammatory mediators involved and discuss their therapeutic potential in the treatment of DME.
Article
Full-text available
Background Improved blood-glucose control decreases the progression of diabetic microvascular disease, but the effect on macrovascular complications is unknown. There is concern that sulphonylureas may increase cardiovascular mortality in patients with type 2 diabetes and that high insulin concentrations may enhance atheroma formation. We compared the effects of intensive blood-glucose control with either sulphonylurea or insulin and conventional treatment on the risk of microvascular and macrovascular complications in patients with type 2 diabetes in a randomised controlled trial. Methods 3867 newly diagnosed patients with type 2 diabetes, median age 54 years (IQR 48-60 years), who after 3 months' diet treatment had a mean of two fasting plasma glucose (FPG) concentrations of 6.1-15.0 mmol/L were randomly assigned intensive policy with a sulphonylurea (chlorpropamide, glibenclamide, or. glipizide) or with insulin, or conventional policy with diet. The aim in the intensive group was FPG less than 6 mmol/L. in the conventional group, the aim was the best achievable FPG with diet atone; drugs were added only if there were hyperglycaemic symptoms or FPG greater than 15 mmol/L. Three aggregate endpoints were used to assess differences between conventional and intensive treatment: any diabetes-related endpoint (sudden death, death from hyperglycaemia or hypoglycaemia, fatal or non-fatal myocardial infarction, angina, heart failure, stroke, renal failure, amputation [of at least one digit], vitreous haemorrhage, retinopathy requiring photocoagulation, blindness in one eye,or cataract extraction); diabetes-related death (death from myocardial infarction, stroke, peripheral vascular disease, renal disease, hyperglycaemia or hypoglycaemia, and sudden death); all-cause mortality. Single clinical endpoints and surrogate subclinical endpoints were also assessed. All analyses were by intention to treat and frequency of hypoglycaemia was also analysed by actual therapy. Findings Over 10 years, haemoglobin A(1c) (HbA(1c)) was 7.0% (6.2-8.2) in the intensive group compared with 7.9% (6.9-8.8) in the conventional group-an 11% reduction. There was no difference in HbA(1c) among agents in the intensive group. Compared with the conventional group, the risk in the intensive group was 12% lower (95% CI 1-21, p=0.029) for any diabetes-related endpoint; 10% lower (-11 to 27, p=0.34) for any diabetes-related death; and 6% lower (-10 to 20, p=0.44) for all-cause mortality. Most of the risk reduction in the any diabetes-related aggregate endpoint was due to a 25% risk reduction (7-40, p=0.0099) in microvascular endpoints, including the need for retinal photocoagulation. There was no difference for any of the three aggregate endpoints the three intensive agents (chlorpropamide, glibenclamide, or insulin). Patients in the intensive group had more hypoglycaemic episodes than those in the conventional group on both types of analysis (both p<0.0001). The rates of major hypoglycaemic episodes per year were 0.7% with conventional treatment, 1.0% with chlorpropamide, 1.4% with glibenclamide, and 1.8% with insulin. Weight gain was significantly higher in the intensive group (mean 2.9 kg) than in the conventional group (p<0.001), and patients assigned insulin had a greater gain in weight (4.0 kg) than those assigned chlorpropamide (2.6 kg) or glibenclamide (1.7 kg). Interpretation Intensive blood-glucose control by either sulphonylureas or insulin substantially decreases the risk of microvascular complications, but not macrovascular disease, in patients with type 2 diabetes. None of the individual drugs had an adverse effect on cardiovascular outcomes. All intensive treatment increased the risk of hypoglycaemia.
Article
Full-text available
S100/calgranulin polypeptides are present at sites of inflammation, likely released by inflammatory cells targeted to such loci by a range of environmental cues. We report here that receptor for AGE (RAGE) is a central cell surface receptor for EN-RAGE (e xtracellular n ewly identified RAGE-binding protein) and related members of the S100/calgranulin superfamily. Interaction of EN-RAGEs with cellular RAGE on endothelium, mononuclear phagocytes, and lymphocytes triggers cellular activation, with generation of key proinflammatory mediators. Blockade of EN-RAGE/RAGE quenches delayed-type hypersensitivity and inflammatory colitis in murine models by arresting activation of central signaling pathways and expression of inflammatory gene mediators. These data highlight a novel paradigm in inflammation and identify roles for EN-RAGEs and RAGE in chronic cellular activation and tissue injury.
Article
Objective: To evaluate the relationship between serum lipid levels, retinal hard exudate, and visual acuity in patients with diabetic retinopathy.Design: Observational data from the Early Treatment Diabetic Retinopathy Study.Participants: Of the 3711 patients enrolled in the Early Treatment Diabetic Retinopathy Study, the first 2709 enrolled had serum lipid levels measured.Main Outcome Measures: Baseline fasting serum lipid levels, best-corrected visual acuity, and assessment of retinal thickening and hard exudate from stereoscopic macular photographs.Results: Patients with elevated total serum cholesterol levels or serum low-density lipoprotein cholesterol levels at baseline were twice as likely to have retinal hard exudates as patients with normal levels. These patients were also at higher risk of developing hard exudate during the course of the study. The risk of losing visual acuity was associated with the extent of hard exudate even after adjusting for the extent of macular edema.Conclusions: These data demonstrate that elevated serum lipid levels are associated with an increased risk of retinal hard exudate in persons with diabetic retinopathy. Although retinal hard exudate usually accompanies diabetic macular edema, increasing amounts of exudate appear to be independently associated with an increased risk of visual impairment. Lowering elevated serum lipid levels has been shown to decrease the risk of cardiovascular morbidity. The observational data from the Early Treatment Diabetic Retinopathy Study suggest that lipid lowering may also decrease the risk of hard exudate formation and associated vision loss in patients with diabetic retinopathy. Preservation of vision may be an additional motivating factor for lowering serum lipid levels in persons with diabetic retinopathy and elevated serum lipid levels.
Article
Objective: To determine whether tight control of blood pressure prevents macrovascular and microvascular complications in patients with type 2 diabetes. Design: Randomised controlled trial comparing tight control of blood pressure aiming at a blood pressure of < 150/85 mm Hg (with the use of an angiotensin converting enzyme inhibitor captopril or a β blocker atenolol as main treatment) with less tight control aiming at a blood pressure of < 180/105 mm Hg. Setting: 20 hospital based clinics in England, Scotland, and Northern Ireland. Subjects: 1148 hypertensive patients with type 2 diabetes (mean age 56, mean blood pressure at entry 160/94 mm Hg); 758 patients were allocated to tight control of blood pressure and 390 patients to less tight control with a medial follow up of 8.4 years. Main outcome measures: Predefined clinical end points, fatal and non-fatal, related to diabetes, deaths related to diabetes, and all cause mortality. Surrogate measures of microvascular disease included urinary albumin excretion and retinal photography. Results: Mean blood pressure during follow up was significantly reduced in the group assigned tight blood pressure control (144/82 mm Hg) compared with the group assigned to less tight control (154/87 mm Hg) (P < 0.0001). Reductions in risk in the group assigned to tight control compared with that assigned to less tight control were 24% in diabetes related end points (95% confidence interval 8% to 38%) (P = 0.0046), 32% in deaths related to diabetes (6% to 51%) (P = 0.019), 44% in strokes (11% to 65%) (P = 0.013), and 37% in microvascular end points (11% to 56%) (P = 0.0092), predominantly owing to a reduced risk of retinal photocoagulation. There was a non-significant reduction in all cause mortality. After nine years of follow up the group assigned to tight blood pressure control also had a 34% reduction in risk in the proportion of patients with deterioration of retinopathy by two steps (99% confidence interval 11% to 50%) (P = 0.0004) and a 47% reduced risk (7% to 70%) (P = 0.004) of deterioration in visual acuity by three lines of the early treatment of diabetic retinopathy study (ETDRS) chart. After nine years of follow up 29% of patients in the group assigned to tight control required three or more treatments to lower blood pressure to achieve target blood pressures. Conclusion: Tight blood pressure control in patients with hypertension and type 2 diabetes achieves a clinically important reduction in the risk of deaths related to diabetes, complications related to diabetes, progression of diabetic retinopathy, and deterioration in visual acuity.
Article
The Diabetes Control and Complications Trial has demonstrated that intensive diabetes treatment delays the onset and slows the progression of diabetic complications in subjects with insulin-dependent diabetes mellitus from 13 to 39 years of age. We examined whether the effects of such treatment also occurred in the subset of young diabetic subjects (13 to 17 years of age at entry) in the Diabetes Control and Complications Trial. One hundred twenty-five adolescent subjects with insulin-dependent diabetes mellitus but with no retinopathy at baseline (primary prevention cohort) and 70 adolescent subjects with mild retinopathy (secondary intervention cohort) were randomly assigned to receive either (1) intensive therapy with an external insulin pump or at least three daily insulin injections, together with frequent daily blood-glucose monitoring, or (2) conventional therapy with one or two daily insulin injections and once-daily monitoring. Subjects were followed for a mean of 7.4 years (4 to 9 years). In the primary prevention cohort, intensive therapy decreased the risk of having retinopathy by 53% (95% confidence interval: 1% to 78%; p = 0.048) in comparison with conventional therapy. In the secondary intervention cohort, intensive therapy decreased the risk of retinopathy progression by 70% (95% confidence interval: 25% to 88%; p = 0.010) and the occurrence of microalbuminuria by 55% (95% confidence interval: 3% to 79%; p = 0.042). Motor and sensory nerve conduction velocities were faster in intensively treated subjects. The major adverse event with intensive therapy was a nearly threefold increase of severe hypoglycemia. We conclude that intensive therapy effectively delays the onset and slows the progression of diabetic retinopathy and nephropathy when initiated in adolescent subjects; the benefits outweigh the increased risk of hypoglycemia that accompanies such treatment. (J PEDIATR 1994;125:177-88)
Article
PURPOSE. To examine the potential of abnormal mfERGs to predict the development of diabetic retinopathy at corresponding retinal locations 1 year later. METHODS. One eye of 11 diabetic patients with nonproliferative diabetic retinopathy (NPDR) and 11 diabetic patients without retinopathy were retested 12 months after initial testing. At each time, mfERGs were recorded from 103 retinal locations, and fundus photographs were taken within 1 month of each recording. Local mfERG implicit times were measured and their z-scores were calculated based on results obtained from 20 age-matched control subjects. mfERG abnormalities were defined as z-scores of 2 or more for implicit time and z-scores of -2 or less for amplitude (P ≤ 0.023). mfERG z-scores were mapped onto fundus photographs, and the relationship between baseline abnormal z-scores and new retinopathy at follow-up was examined. RESULTS. New retinopathy developed in 7 of the eyes with NPDR after 1 year. In these eyes, 70% of the mfERGs in areas of new retinopathy had abnormal implicit times at baseline. In contrast, only 24% of the responses in regions that remained retinopathy free were abnormal at baseline. Relative risk of development of new retinopathy over 1 year in the areas with abnormal baseline mfERG implicit times was approximately 21 times greater than that in the areas with normal baseline mfERGs (odds ratio = 31.4; P < 0.001). Eyes without initial retinopathy did not develop new retinopathy within the study period, although 4 of these 11 eyes had abnormal implicit times at baseline. mfERG implicit times tended to be more delayed at follow-up than at baseline in NPDR eyes, but not in eyes without retinopathy and control eyes. mfERG amplitudes had no predictive power. CONCLUSIONS. Localized functional abnormalities of the retina reflected by mfERG delays often precede the onset of new structural signs of diabetic retinopathy. Those functional abnormalities predict the local sites of new retinopathy observed 1 year later.
Article
Flat preparations of the retina afford an unusually favorable opportunity to study blood vessel architecture (Ballantyne and Lowenstein1). The retinal vessels have been visualized in the past by the following methods: 1. injection of India ink into the heart, major arteries, or directly into the retinal artery (Michaelsen,2 Ashton,3 Keeney and Barlow,4 and others); 2. latex injection into the aorta (Janes5); 3. benzidine-peroxidase staining of the red blood cells (Michaelsen2); 4. infusion of silver nitrate with subsequent reduction of the silver to outline the reticular fibers,6 and 5. staining of the full thickness of
Article
purpose. A critical event in the pathogenesis of diabetic retinopathy is the inappropriate adherence of leukocytes to the retinal capillaries. Advanced glycation end-products (AGEs) are known to play a role in chronic inflammatory processes, and the authors postulated that these adducts may play a role in promoting pathogenic increases in proinflammatory pathways within the retinal microvasculature. methods. Retinal microvascular endothelial cells (RMECs) were treated with glycoaldehyde-modified albumin (AGE-Alb) or unmodified albumin (Alb). NFκB DNA binding was measured by electromobility shift assay (EMSA) and quantified with an ELISA. In addition, the effect of AGEs on leukocyte adhesion to endothelial cell monolayers was investigated. Further studies were performed in an attempt to confirm that this was AGE-induced adhesion by co-incubation of AGE-treated cells with soluble receptor for AGE (sRAGE). Parallel in vivo studies of nondiabetic mice assessed the effect of intraperitoneal delivery of AGE-Alb on ICAM-1 mRNA expression, NFκB DNA-binding activity, leukostasis, and blood-retinal barrier breakdown. results. Treatment with AGE-Alb significantly enhanced the DNA-binding activity of NFκB (P = 0.0045) in retinal endothelial cells (RMECs) and increased the adhesion of leukocytes to RMEC monolayers (P = 0.04). The latter was significantly reduced by co-incubation with sRAGE (P < 0.01). Mice infused with AGE-Alb demonstrated a 1.8-fold increase in ICAM-1 mRNA when compared with control animals (P < 0.001, n = 20) as early as 48 hours, and this response remained for 7 days of treatment. Quantification of retinal NFκB demonstrated a threefold increase with AGE-Alb infusion in comparison to control levels (AGE Alb versus Alb, 0.23 vs. 0.076, P < 0.001, n = 10 mice). AGE-Alb treatment of mice also caused a significant increase in leukostasis in the retina (AGE-Alb versus Alb, 6.89 vs. 2.53, n = 12, P < 0.05) and a statistically significant increase in breakdown of the blood-retinal barrier (AGE Alb versus Alb, 8.2 vs. 1.6 n = 10, P < 0.001). conclusions. AGEs caused upregulation of NFκB in the retinal microvascular endothelium and an AGE-specific increase in leukocyte adhesion in vitro was also observed. In addition, increased leukocyte adherence in vivo was demonstrated that was accompanied by blood-retinal barrier dysfunction. These findings add further evidence to the thinking that AGEs may play an important role in the pathogenesis of diabetic retinopathy.
Article
Purpose Retinopathy is the most common microvascular complication of diabetes. The clinicopathology of microvascular lesions and neuroglial dysfunction in the diabetic retina have been extensively studied, although the relative contribution of various biochemical sequelae of hyperglycaemia remains ill‐defined. The formation and accumulation of advanced glycation endproducts (AGEs) is an important pathogenic pathway in the progression of diabetic retinopathy although some of the cellular and molecular pathologies initiated by these adducts in retinal cells remain unknown. Methods This presentation will cover several aspects of AGE‐linked retinal pathology and demonstrate opportunities for therapeutic intervention. The studies outlined will cover a wide range of molecular cell biology approaches using appropriate in vitro and in vivo model systems. Results It will be demonstrated that AGEs form in vivo in the diabetic retina through the reaction of alpha‐oxaloaldehydes leading to significant modifications of retinal proteins. Evidence will be presented to demonstrate that these AGEs act as significant effectors of retinal vascular and neuroglial cell dysfunction, leading to pro‐inflammatory responses, growth factor imbalance and, ultimately, neurovascular lesions such as blood retinal barrier dysfunction and microvascular degeneration. The protective role of novel AGE‐inhibitors will also be shown. Conclusion Evidence now points towards a pathogenic role for advanced glycation in the initiation and progression of diabetic retinopathy and this review lecture will outline the current state of knowledge of AGE‐related pathology in the retina at a cellular and molecular level.