You are on page 1of 8

Perspectives

AGEs and Diabetic Retinopathy


Alan W. Stitt

The pathogenesis of diabetic retinopathy is multifactorial, and The DCCT (Diabetes Control and Complications Trial) and
a range of hyperglycemia-linked pathways have been impli- UKPDS (UK Prospective Diabetes Study) population studies in
cated in the initiation and progression of this condition. All type 1 and type 2 diabetic patients, respectively, have estab-
cells in the retina are affected by the diabetic milieu, and in lished the relationship between hyperglycemia and retinopa-
view of such disease and tissue complexity, it is unlikely that thy.10,11 These seminal studies, and many others, point toward
any single process is solely responsible for retinal pathophysi- hyperglycemia as being critical in the pathogenesis, although it
ology. Nevertheless, establishing causal mechanisms remains often occurs in unison with dyslipidemia and hypertension.
an important research goal. This review concentrates on the This epidemiology provides the foundation for ongoing re-
formation of advanced glycation end products (AGEs) and the search, seeking to identify the cellular and molecular mecha-
role they play in diabetic retinopathy. Perspective is provided nisms that underpin diabetic retinopathy. The formation of
on advanced glycation in the retina, the impact that this pro- advanced glycation end products (AGEs) and the activation of
cess has on retinal cell function, and how it relates to other receptors for AGEs are the focus of this article, although it
pathogenic pathways. Emphasis is also placed the modulatory should be appreciated that hyperglycemia can simultaneously
role of the receptor for AGEs (RAGE) and how its activation provoke a range of other pathogenic mechanisms in retinal
could evoke retinal inflammatory disease. Further research is cells in vitro and in vivo.12 Such pathways to diabetic retinop-
needed to achieve a clear understanding of the cellular and athy should not necessarily be viewed as independent phenom-
molecular processes that underpin diabetic retinopathy’s initi- ena. Brownlee et al.13 have proposed a unifying concept
ation and progression. Such advances in basic mechanisms may whereby hyperglycemia increases superoxide production (via
lead to effective treatments that can prevent progression of the mitochondrial electron transport chain) which in turn
retinopathy from the point of the diagnosis of diabetes to initiates accelerated AGE formation and also exacerbates inter-
sight-threatening proliferative diabetic retinopathy (PDR) and related pathogenic responses. This hypothesis has been rein-
diabetic macular edema (DME). (Invest Ophthalmol Vis Sci. forced in the field of retinopathy, in which three biochemical
2010;51:4867– 4874) DOI:10.1167/iovs.10-5881 abnormalities involving AGE formation, flux through the hex-
osamine pathway and diacylglycerol-mediated activation of
R etinopathy is the most common microvascular complica-
tion of diabetes, and it remains a major cause of visual
impairment worldwide.1 Epidemiology studies indicate that, at
PKC-␤ can be attenuated with benfotiamine. This vitamin B1
thiamine derivative stimulates transketolase activity and shunts
excess triose phosphates toward the reductive pentose phos-
20 years after diagnosis, most patients with type 1 diabetes will phate pathway, which is impaired in high-glucose diabetes.14
experience retinopathy, whereas ⬃80% of insulin-dependent By converging on three harmful pathways, benfotiamine pre-
patients with type 2 diabetes and ⬃50% of patients with non– vents high-glucose–induced dysfunction in retinal microvascu-
insulin-dependent type 2 diabetes will have retinopathy.2 lar cells,15 whereas treatment of diabetic animals protects
Microvascular lesions, such as microaneurysms, blood bar- against the key lesions of retinopathy.16
rier dysfunction, and capillary dropout, are key features of
diabetic retinopathy.3,4 However, it should be appreciated that
the sole purpose of the retinal circulation is to support the BIOCHEMISTRY OF AGE FORMATION
metabolic demands of the inner retinal neurons and glia and
that these cells are also damaged appreciably during diabetes.
How AGEs Are Formed
Such neuronal and glial dysfunction occurs in unison with Nonenzymatic-glycation reactions between reducing sugars
blood flow abnormalities and often before the appearance of and the free amino groups on proteins, lipids, and DNA are an
overt microvascular damage.5 In diabetic patients and short- inevitable consequence of aldehyde reactivity. As a conse-
term animal models of diabetes, reversible retinal psychophys- quence, many proteins in vivo carry some burden of chemi-
ical and electrophysiological alterations are evident.6,7 Such cally attached carbohydrate. An understanding of this chemis-
early-stage alterations may be signposts for the irreversible try was established in 1912 by the food chemist, Louis Camille
retinal microvascular, neuronal, and glial damage observed Maillard, who reported formation of brown products upon
after longer term diabetes.8,9 heating mixtures of amino acids and sugars. The Maillard, or
browning, reaction begins with the formation of a Schiff base
between glucose and ␧-amino groups that slowly rearranges to
From the Centre for Vision and Vascular Science, Queens Univer- relatively stable Amadori adducts, an example of which is
sity Belfast, Northern Ireland, United Kingdom. hemoglobin A1c (HbA1c). Both the Schiff base and the Amadori
Supported by the Juvenile Diabetes Research Foundation (JDRF), product can undergo further oxidation and dehydration with
Fight for Sight (UK), Action Medical Research, and the Medical Re- concentrations ultimately dependent on both forward and re-
search Council. verse reactions. The forward reactions give rise to additional
Submitted for publication May 13, 2010; revised July 9, 2010; irreversible protein-bound compounds collectively termed ad-
accepted July 9, 2010.
Disclosure: A.W. Stitt, None
vanced glycation end products (AGEs). During diabetes, the
Corresponding author: Alan W. Stitt, Centre for Vision and rate of formation of AGEs exceeds that predicted by first-order
Vascular Science, Queen’s University Belfast, Royal Victoria Hospi- kinetics. Thus, over time, even modest hyperglycemic excur-
tal, Grosvenor Road, Belfast, BT12 6BA, Northern Ireland, UK; sions can result in significant adduct accumulation on long-
a.stitt@qub.ac.uk. lived macromolecules.17

Investigative Ophthalmology & Visual Science, October 2010, Vol. 51, No. 10
Copyright © Association for Research in Vision and Ophthalmology 4867
4868 Stitt IOVS, October 2010, Vol. 51, No. 10

AGE adducts are highly stable at physiological pH, and their ALEs May Play an Important Role in
rate of accumulation in tissues depends on factors such as Retinal Disease
availability of metal ions, redox balances, and longevity of the
modified protein. Although AGEs may lead to pigmentation or Dyslipidemia is often overlooked as a pathogenic force in
fluorescence, they can also inflict considerable damage on diabetic retinopathy.35 In the context of this review, lipid
proteins through cross-linking, changing tertiary structure, peroxidation reactions can also form a class of Maillard pro-
conferring resistance to digestion, altering enzymatic activity, ducts, the ALEs, which are linked to diabetes and dyslipide-
or impairing receptor recognition. AGE adducts form from mia.36 ALEs form through the lipoperoxidative production of
many different precursors that contribute to the heterogeneity reactive aldehyde species. Among the best characterized are
of these chemical structures. Numerous AGE adducts have N ␧ -(2-propenal)lysine and dihydropyridine-type adducts
been identified in vivo, including; N␧-(carboxymethyl) lysine (malondialdehyde-derived), hemiacetal and pyrrole adducts
(CML), crossline, pentosidine, furoyl-furanyl imidazole (FFI), hy- (4-hydroxy-2-nonenal, and 4-hydroxyhexenal-derived), and
droimidazolone, argpyrimidine, glyoxal lysine dimer (GOLD), and N␧-(3-formyl-3,4-dehydropiperidino)lysine, or FDP-lysine; ac-
methylglyoxal lysine dimer (MOLD).18 rolein-derived).37 Recent work has also shown that hemo-
globin levels of the ACR-derived ALE, FDP-lysine, are asso-
ciated with the severity of retinopathy in patients with type
AGEs Form from a Range of Precursors 1 or type 2 diabetes.38 Clearly, ALEs represent an important
Glucose is often viewed as the principal AGE precursor; how- source of protein modification, especially in lipid-rich,
ever, it is considerably less reactive than ␣-oxaloaldehydes highly oxidative environments, such as that in the retina.
such as glyoxal (GO), methylglyoxal (MGO), and 3-deoxyglu- The understanding of the role of ALEs in diabetic retinopa-
cosone (3-DG), which arise from glycolytic metabolism and thy lags far behind that which is known about AGEs, and
can form AGEs very rapidly.19,20 For example, GO reacts with these adducts therefore warrant further study.
arginine residues to form carboxymethyl-arginine (CMA),21
whereas MGO can give rise to the AGEs N␧-(carboxyethyl) PATHOGENIC ROLE OF AGES
lysine (CEL) and arginine-hydroimidazolone.19,22 The concen- IN DIABETIC RETINOPATHY
trations of these reactive carbonyls rise in high-glucose– ex-
posed cells and occur at elevated levels in diabetic serum, and AGEs and Clinical Correlation
they constitute the major source of AGEs in vivo.22,23 Indeed, with Diabetic Retinopathy
in diseases of carbonyl stress, such as diabetic nephropathy,
the AGEs can reach exceptionally high levels.24 AGEs affect cells from three main perspectives: as adducts
occurring on modified serum proteins, as endogenous adducts
formed as a consequence of glucose metabolism, or as extra-
AGEs Are Also Derived from Food cellular matrix–immobilized modifications on long-lived struc-
AGEs and ALEs are abundant in food. Indeed a typical Western tural proteins (Fig. 1). All these AGEs can be analyzed in serum,
diet contains high levels of fat and sugar and, being highly cells, and tissues using analytical and/or immunocytochemical
thermally processed, can lead to high levels of harmful ad- approaches. Clinically, most quantification in patient tissues
ducts.25 Consumption of a conventional Western diet typically and serum has been based on ELISA of a range of AGE-antibod-
leads to a daily intake of ⬃25 to 75 mg AGE/ALEs (advanced ies, but this method has often produced confounding out-
lipoxidation end products),25 and a proportion of these ad- comes. With this proviso, patient-based studies have revealed
ducts pass the gut epithelium and can appear as plasma that the levels of AGEs in serum correlate with the clinical
AGEs.26 Food-derived AGEs may be linked to vascular dysfunc- progression of diabetic retinopathy.39 Although many reports
tion, especially in situations in which there is renal dysfunction have measured a range of ill-defined AGE moieties, others have
and poor clearance of plasma AGEs.27,28 The potential involve- used adduct-specific antibodies or chemical analysis for CML,
ment of food-derived AGEs and ALEs in diabetic retinopathy pentosidine, or hydroimidazolone40 – 42 and also have found
has not been studied. association with diabetic retinopathy. It should be acknow-
ledged that some reports have demonstrated no correlation
between AGE levels and retinopathy in diabetic patients,40,43
Detoxification Systems Can Limit AGE Formation although the apparent disparity may be related to variations in
In Vivo patient populations, the presence of nephropathy, and/or the
Cells have evolved systems that provide endogenous protec- nonuniformity of assays for AGE quantification.
tion against dicarbonyls, and several detoxifying enzymes have
been identified. For example, a glutathione-dependent glyox-
AGEs as Robust Biomarkers for Disease Risk
alase complex (formed from glyoxalase I [GLO1] and glyox- AGE-modified proteins are readily cleared from the blood-
alase II [GLO2] components) acts as a detoxification system for stream (except during renal dysfunction), and thus quantifica-
GO and MGO, which are converted to D-lactate.29 Endothelial tion of these adducts in serum may not always provide robust
cells transfected to overexpress GLO1 accumulate less MGO- biomarkers for disease. By contrast, AGE modification of extra-
derived AGEs30 and are protected against high-glucose–in- cellular matrix isolated from skin biopsies often provides more
duced responses.31 The GLO1 detoxification system has been meaningful data,44 illustrated by the DCCT skin collagen ancil-
shown to be critical for retinal pericyte survival, but this may lary study group.45 They demonstrated that cross-linked AGEs
be insufficient during diabetes, since these cells undergo apo- on long-lived skin proteins are significantly associated with the
ptosis as a direct result of MGO-derived AGE formation.32 The progression of diabetic retinopathy. More than 200 patients
importance of this enzyme system is supported by findings in from the original DCCT were followed up for a further 10 years
studies in which Caenorhabditis elegans was engineered to under the auspices of the Epidemiology of Diabetes Interven-
overexpress GLO1. These worms contain fewer AGEs and tions and Complications (EDIC) Trial.46 The results revealed
show a significantly increased lifespan when compared with that levels of diabetic retinopathy were significantly lower in
wild-type counterparts.33,34 Clearly, there is a potential for the group initially maintained under tight glycemic control
harnessing the detoxifying property of enzymes such as GLO1, and that these benefits extended far beyond the period of
to protect against diabetic retinopathy. intensive insulin therapy.46 The patients under conventional
IOVS, October 2010, Vol. 51, No. 10 AGEs and Diabetic Retinopathy 4869

FIGURE 1. AGEs interact with cells N2-Carboxymethyl-lysine


from three main routes. Route 1: O C
AGE-modified serum proteins, such CH (CH )
2 4 2

NHCH CO
2
H N
as CML, may interact with vascular RAGE
endothelium via AGE receptors such
as RAGE, which can activate NF-␬B Increased expression of
transcription leading to enhanced ICAM, VCAM, E-selectin,
VEGF, RAGE, TNF- ROS NO
expression of adhesion molecules
and secretion of cytokines such as JAK
TNF- ␣ and VEGF. Serum-derived 1
Rac1 ERK1/2
AGEs reach pericytes via transendo- Glucose 2
thelial trafficking or as a result of Glucose-6-P

blood–retinal barrier dysfunction. Nuclear


Fructose-6-P
Methylglyoxal
transcription
These serum AGEs may also inter- factors Glyceraldehyde-3-P O
+
NAD
act directly with cell surface glyco- GAPDH O
HC
C
C
O
AP1/NF- B/ NADH
2
3
proteins with potentially damaging STAT
H

effects on membrane integrity and


function. Route 2: AGEs can form
directly from reaction of glucose
with amino groups, but this sugar is
much less reactive than carbonyls,
such as methylglyoxal. Methylg-
lyoxal can arise by spontaneous ␤
elimination of phosphate from tri- O C N
Pentosidine
ose phosphates, the concentrations crosslink CH (CH )
2 8
NH+
H N HN N
of which are increased during hy- (fluorophore) C O 3
(CH ) HC
perglycemia because of the in- 2 4
N H ? ? ?
creased flux of glucose through gly-
colysis. Route 3: Sedentary cells like endothelium encounter AGEs such as pentosidine-derived cross-links within basement membrane
proteins where they may influence cell attachment through disruption of integrin signaling. Integrins of various subunit combinations are
important for function and ongoing survival cues and disruption of signaling contributes to dysfunction and death. There are many
uncertainties about how the AGE adducts immobilized in the basement membrane could interact with RAGE (or other AGE-receptors)
expressed on the basal plasma membrane, but this effect may be a significant mode of cell dysfunction.

control for the first 10 years maintained a hyperglycemic or (as a function of disruption of the glutamate transporter63)
metabolic memory and retained a strong association with ret- which may contribute to excitotoxicity in retinal neurons.64
inopathy progression. CML-modified skin collagen predicted Significantly, Winkler et al.65 have demonstrated that Müller
the progression of retinopathy (and nephropathy), even after cells exposed to high glucose conditions in vitro produce
initiation of intensive insulin therapy.46 Furthermore, the pre- excess lactate, indicative of increased glycolytic flux,65 and this
dictive effect of hemoglobin A1c (HbA1c) vanished after ad- effect leads to greater production of MGO. Hypoxia is also
justment for AGEs, suggesting that accumulation of these ad- known to enhance glycolytic metabolism through increased
ducts on long-lived protein is an excellent marker for HIF-1␣-dependent expression of glycolytic enzymes in the con-
retinopathy risk and could offer a molecular-based explanation version of cells to a predominantly glycolytic metabolic state in
for the metabolic memory phenomenon.17 the absence of oxygen (the Pasteur effect). Hypoxia, by in-
creasing glycolysis and MGO synthesis, could lead to significant
AGEs in the Diabetic Retina AGE formation in the diabetic retina.
AGEs have been extensively quantified in various ocular tissues
and are often elevated during ageing and in diabetic subjects AGEs Evoke Retinal Oxidative Stress and Retinal
when compared to nondiabetic control subjects.47 This in- Cell Death
cludes vitreous collagen,48 where the AGE levels correlate with
diabetic retinopathy.49 In the diabetic retina, AGEs and/or late Oxidative stress results from the disequilibrium between pro-
Amadori products have been localized to vascular cells, neu- and antioxidants in biological systems,66 and this pathway is
rons, and glia.50 –55 This would be expected to have patho- intimately linked to the formation of AGEs.13,67,68 Numerous
genic implications for the individual cells and retinal function. studies have reported that oxidative stress is increased in dia-
Although differential accumulation of AGEs exists in the retina betic patients and that it plays an important role in the patho-
over the course of life, diabetes significantly enhances the genesis of diabetic complications, including retinopathy.69,70
occurrence of these adducts in the vascular and neural tissue Previous work has demonstrated that the concentration of
components.54 superoxide is elevated in the retina of diabetic rats and in
It has been demonstrated that MGO-derived hydroimida- retinal cells incubated in high-glucose media.61,71,72
zolone is increased 279% in 24-week diabetic rat retina,56 and Of importance, it has been shown that inhibition of super-
this finding emphasizes the fact that MGO could be the major oxide with antioxidants73,74 or overproduction of mitochon-
source of AGEs in this tissue. In terms of cell localization for drial superoxide dismutase (SOD)75 can protect against capil-
AGEs, many adducts occur at high levels in the Müller macro- lary degeneration during diabetic retinopathy in experimental
glia, and these increase as diabetes progresses.57 This fact is diabetes, although how this influences AGE accumulation in
significant, because Müller glia have a unique role in the archi- the retina has not been studied. Retinal capillary degeneration
tecture and physiology of the retina and show considerable remains a hallmark of retinopathy in diabetic animal models
dysfunction during the hyperglycemia and hypoxia experi- and patients76 and these vessels seem to be important targets
enced by diabetic retina.58,59 This dysfunction is manifested by for both AGE- and superoxide-induced toxicity.77 For example,
increased expression of glial fibrillary acidic protein (GFAP),60 AGEs induce toxic effects on retinal pericytes by causing oxi-
NO production,61,62 and concomitant synthesis of glutamate dative stress and subsequent apoptosis.78 In addition, some
4870 Stitt IOVS, October 2010, Vol. 51, No. 10

studies have indicated that AGEs cause osteoblastic differenti- INFLAMMATION, RAGE, AND DIABETIC RETINOPATHY
ation and calcification in retinal pericytes by the activation of
alkaline phosphatases.79 Pericytes growing on AGE-modified Inflammatory Processes in Diabetic Retinopathy
basement membrane show acute attenuation of endothelin-1
The involvement of inflammatory processes in the initiation of
(ETA receptor–mediated) contraction, suggesting that AGE
neurovascular lesions during diabetic retinopathy has received
cross-linking in a surrounding matrix significantly influences
recent attention. Global mRNA expression profiling has high-
pericyte physiology.80 Indeed, longer exposure times to these
lighted altered expression of proinflammatory cytokines and
substrate AGEs induce loss of integrin signaling and apoptosis.81 interrelated pathways, not only in the retinal vessels,100 but
Retinal microvascular endothelial cells also show proangio- also in the neuroglia.101 There is undoubtedly a complex mi-
genic responses to AGEs at lower concentrations by the in- lieu of dysregulated proinflammatory factors apparent in dia-
volvement of MAPK, PKC, and NF-␬B signaling pathways,82 betic retina, but there is strong evidence of involvement of
although at higher concentrations, these adducts are toxic to major mediators of inflammation such as IL-1␣, IL-1␤, and IL-6
endothelial cells83 and in vivo may eventually lead to enhanced and TNF␣102–104 that may be linked to microglial activation
microvascular closure.84 Under hyperglycemic conditions, ret- and infiltrating monocytes that are increased in diabetic retina,
inal microvascular endothelial cells accumulate MGO and both in humans105,106 and in animal models.107,108
MGO-derived AGE adducts (such as hydroimidazolone and arg-
pyrimidine) which in vivo contribute to premature closure of RAGE as a Component of the Innate
capillaries.85 AGEs cause upregulation of ICAM, which medi- Immune Response
ates retinal capillary leukocyte adherence and inner blood–
retinal barrier breakdown.86 Independent of the complexities The receptor for AGEs (RAGE) is the most established AGE-
of the diabetic milieu, nondiabetic mice exposed to diabetic- binding protein and acts as a signaling receptor for at least
like levels of injected AGE-albumin show increased retinal two distinct AGEs: CML109 and hydroimidazolone adducts.18
expression of VEGF concomitant with blood–retinal barrier RAGE is now known to be a key component of the innate
dysfunction.87 Similar treatments may cause loss of pericytes,88 immune response110 and binds to multiple ligands including
and the evidence suggests that high serum levels of AGE- S100B, high-mobility group box (HMGB)-1, amyloid-␤, and
modified proteins (as particularly evident in diabetic patients Mac-1 (CD11b/CD18).111,112
with renal dysfunction) induce lesions that are comparable to RAGE is constitutively expressed in a range of tissues, such
those that occur during diabetic retinopathy. as brain, kidney, liver, heart, and the vasculature, and in various
cell types, including neurons, endothelium, smooth muscle,
epithelium, and inflammatory cells.113 RAGE and downstream
AGE Inhibition and Prevention of Retinopathy proinflammatory signaling on ligand-binding are associated
with several disease states such as diabetic complications,
A pharmacologic strategy for AGE-inhibition commenced with Alzheimer’s disease, cancer, and viral infections.110,112,114,115
the small nucleophilic hydrazine compound called aminogua- As a result of alternative mRNA splicing and proteolytic cleav-
nidine (or pimagedine).89 This drug is a potent inhibitor of age RAGE exists in several forms. The best known is soluble
AGE-mediated cross-linking and has been shown to prevent (s)RAGE, which is composed of the extracellular domains but
diabetic vascular complications, including diabetic retinopa- lacks the transmembrane and cytosolic sequences. sRAGE may
thy, in experimental animals.50,90 –93 Aminoguanidine has been act as a dominant negative isoform and block RAGE signaling
evaluated in a multicenter clinical trial where it failed to by function as an extracellular “decoy receptor” to inhibit
achieve statistically significant lowering of serum creatinine, RAGE ligand-binding.116
and urinary albumin but showed a positive trend toward slow- RAGE dimerization occurs on ligand binding117 and, in
ing the progression of overt nephropathy and retinopathy.94 association with a binding protein diaphanous (Dia)-1,118 in-
However, it is now known that aminoguanidine is not a spe- tracellular signaling is initiated. This process can result in
cific AGE inhibitor and also acts as an effective iNOS inhibi- phosphorylation of various protein kinases involving MAPKs,
tor.95 Rac/Cdc42, and JAK/STATs and subsequently activate the
Agents with post-Amadori product scavenging properties NF-␬B pathway.119,120 This signal transduction links RAGE to
prevent experimental diabetic retinopathy. The so-called Ama- several inflammation-related cell responses, such as apoptosis,
dorins have an ability to scavenge reactive carbonyls and there- mobility, migration, and proinflammatory gene expression.116
fore inhibit the conversion of Amadori intermediates to AGEs Thus, RAGE has been the focus for several small-molecule
and ALEs.96 The derivative of vitamin B6, pyridoxamine (Pyri- drugs or neutralizing antibodies that can regulate ligand bind-
dorin; NephroGenex, Princeton, NJ) is an efficacious and spe- ing or downstream signal transduction and thereby prevent
cific post-Amadori inhibitor97 that reduces retinal AGE accu- disease.121
mulation and attenuates the upregulation of basement
membrane–associated genes and capillary acellularity in dia- RAGE Involvement in Diabetic Retinopathy
betic rat retina.57 More recently, an agent called LR-90 has been In the retina, RAGE expression has been predominantly local-
developed as an effective multistage inhibitor of both AGE/ALE ized to glia in the inner retina, and this receptor appears to be
formation with associated renoprotective and anti-inflamma- upregulated in diabetic conditions.122 AGE-RAGE ligands have
tory potential.98 LR-90 prevents diabetic retinopathy in rats at also been demonstrated in the retina, and they often occur at
doses that are several-fold less than the doses of pyridoxamine.99 higher levels during diabetes.51,57,87 Other RAGE ligands in-
For many patients there will have been extensive AGE cluding S100/calgranulins and HMGB1 are evident in the vit-
formation at the time of type 2 diabetes diagnosis. Therefore, it reous and preretinal membranes of eyes with proliferative
would be beneficial to attack established cross-links in tissues diabetic retinopathy (PDR) and proliferative vitreoretinopa-
and enable subsequent renal clearance of peptide fragments. thy (PVR).123 Hyperglycemic mice exhibit enhanced RAGE
An AGE cross-link breaker attacks AGE-derived protein cross- expression in the inner retina, particularly in Müller cells,
links and treatment with this drug reduces vascular stiffening which show elevated receptor levels at the vitreoretinal sur-
in experimental diabetes.90 The effects of AGE breakers on face.124 This finding opens a further new paradigm for possible
diabetic retinopathy have yet to be evaluated. RAGE-mediated involvement in retinal neuropathic abnormali-
IOVS, October 2010, Vol. 51, No. 10 AGEs and Diabetic Retinopathy 4871

ties during diabetes, and recent studies have indicated a role 3. Curtis TM, Gardiner TA, Stitt AW. Microvascular lesions of dia-
for the receptor in Müller cell dysfunction.125 It has been betic retinopathy: clues towards understanding pathogenesis?
reported that hyperglycemia increases RAGE, S100A8, Eye (Lond.). 2009;23:1496 –1508.
S100A12, and HMGB1 expression in human aortic endothelial 4. Frank RN. Diabetic retinopathy. N Engl J Med. 2004;350:48 –58.
cells, a response that can be normalized by a superoxide 5. Antonetti DA, Barber AJ, Bronson SK, et al. Diabetic retinopathy:
dismutase mimetic.126 We have recently shown that this re- seeing beyond glucose-induced microvascular disease. Diabetes.
sponse occurs in the diabetic retina whereby hyperglycemia in 2006;55:2401–2411.
vivo and HG exposure to Müller cells in vitro induce the 6. Hancock HA, Kraft TW. Oscillatory potential analysis and ERGs of
normal and diabetic rats. Invest Ophthalmol Visual Sci. 2004;45:
expression of RAGE and its ligands, leading to cytokine signal-
1002–1008.
ing. This signaling, in turn, leads to RAGE signaling and proin-
7. Jackson GR, Owsley C. Visual dysfunction, neurodegenerative
flammatory cytokine expression, a response that further impli- diseases, and aging. Neurol Clin. 2003;21:709 –728.
cates involvement of this receptor in retinal inflammatory 8. Martin PM, Roon P, Van Ells TK, Ganapathy V, Smith SB. Death of
disease.127 retinal neurons in streptozotocin-induced diabetic mice. Invest
Ophthalmol Visual Sci. 2004;45:3330 –3336.
RAGE Blockade as a Potential Therapeutic 9. Barber AJ, Lieth E, Khin SA, Antonetti DA, Buchanan AG, Gardner
Strategy for Diabetic Retinopathy TW. Neural apoptosis in the retina during experimental and
human diabetes: early onset and effect of insulin. J Clin Invest.
sRAGE can prevent Müller cell dysfunction125 during diabetes 1998;102:783–791.
and retinal capillary leukostasis in AGE-infused normal mice.86 10. DCCT. Hypoglycemia in the Diabetes Control and Complications
The clinical potential for reducing RAGE signaling in the dia- Trial. The Diabetes Control and Complications Trial Research
betic retina is further underscored by development of new Group. Diabetes. 1993;46:271286.
RAGE-regulating agents. One such agent is TTP488, which is an 11. (UKPDS) UPDS. Effect of intensive blood-glucose control with
orally delivered small molecule, for which phase II studies have metformin on complications in overweight patients with type 2
been completed in patients with Alzheimer’s disease. Use of diabetes (UKPDS 34). Lancet. 1998;352:854 – 865.
these agents and other similar strategies is exciting, because 12. Madsen-Bouterse SA, Kowluru RA. Oxidative stress and diabetic
they directly address RAGE-ligand binding and show great retinopathy: pathophysiological mechanisms and treatment per-
potential for diabetic complications. Evaluation of the role of spectives. Rev Endocr Metab Disord. 2008;9:315–327.
RAGE inhibition in diabetic retinopathy is ongoing in several 13. Brownlee M. Biochemistry and molecular cell biology of diabetic
laboratories. complications. Nature. 2001;414:813– 820.
Some commonly used diabetes drugs, such as thiazolidine- 14. Thornalley PJ. The potential role of thiamine (vitamin B1) in
diabetic complications. Curr Diabetes Rev. 2005;1:287–298.
diones (e.g., rosiglitazone),128 or calcium channel blockers, such
15. Berrone E, Beltramo E, Solimine C, Ape AU, Porta M. Regulation
as nifedipine,129 have been shown to reduce RAGE expression in
of intracellular glucose and polyol pathway by thiamine and
endothelial cells and could serve to limit the proinflammatory benfotiamine in vascular cells cultured in high glucose. J Biol
effects of AGEs. In addition, angiotensin II receptor blockers Chem. 2006;281:9307–9313.
(ARBs) can reduce RAGE expression, and this effect should be 16. Hammes HP, Du X, Edelstein D, et al. Benfotiamine blocks three
factored into the benefit that these agents have in preventing major pathways of hyperglycemic damage and prevents experi-
diabetic retinopathy.130 In addition to the downregulation of mental diabetic retinopathy. Nat Med. 2003;9:294 –299.
RAGE and associated reduction in oxidative stress, ARBs can act as 17. Monnier VM, Sell DR, Genuth S. Glycation products as markers
selective regulators of PPAR␥, which suggests some cross-talk and predictors of the progression of diabetic complications. Ann
between the AGE–RAGE axis and PPAR␥ modulation.131 Thus, a N Y Acad Sci. 2005;1043:567–581.
complex interplay between enhanced levels of AGEs, suppressed 18. Goldin A, Beckman JA, Schmidt AM, Creager MA. Advanced
antioxidant status and upregulation of the RAGE axis may to- glycation end products: sparking the development of diabetic
gether play a pivotal role in the progression of diabetic retinopa- vascular injury. Circulation. 2006;114:597– 605.
thy. 19. Thorpe SR, Baynes JW. Maillard reaction products in tissue
proteins: new products and new perspectives. Amino Acids.
2003;25:275–281.
CONCLUSION 20. Lal S, Szwergold BS, Taylor AH, et al. Metabolism of fructose-3-
phosphate in the diabetic rat lens. Arch Biochem Biophys. 1995;
Long-term management of retinopathy in the ever-expanding 318:191–199.
diabetic patient population involves precise regulation of gly- 21. Glomb MA, Pfahler C. Amides are novel protein modifications
cemic, vasotensive, and lipidemic profiles, most effectively in formed by physiological sugars. J Biol Chem. 2001;276:41638 –
combination with drugs that ameliorate an array of biochemi- 41647.
cal and metabolic abnormalities. Early-stage experimental work 22. Thornalley PJ, Langborg A, Minhas HS. Formation of glyoxal,
methylglyoxal and 3-deoxyglucosone in the glycation of proteins
suggests that AGE formation and activation of AGE-receptors
by glucose. Biochem J. 1999;344:109 –116.
represent important, interconnected pathogenic mechanisms
23. Kilhovd BK, Giardino I, Torjesen PA, et al. Increased serum levels
in diabetic retinopathy, and inhibition of these pathways pre- of the specific AGE-compound methylglyoxal-derived hydroimi-
sents a valid avenue for therapeutic exploitation. As our knowl- dazolone in patients with type 2 diabetes. Metabolism. 2003;52:
edge of Maillard chemistry and its biological impact improves, 163–167.
alongside elucidation of AGE-receptor signaling in various ret- 24. Miyata T, Ishikawa N, van Ypersele de Strihou C. Carbonyl stress
inal cell-types, there may be exciting new opportunities for and diabetic complications. Clin Chem Lab Med. 2003;41:1150 –
therapeutic management of diabetic retinopathy at all stages of 1158.
the disease. 25. Henle T. AGEs in foods: do they play a role in uremia? Kidney Int
Suppl. 2003;S145–S147.
References 26. Koschinsky T, He CJ, Mitsuhashi T, et al. Orally absorbed reactive
glycation products (glycotoxins): an environmental risk factor in
1. Marshall SM, Flyvbjerg A. Prevention and early detection of vas- diabetic nephropathy. Proc Natl Acad Sci U S A. 1997;94:6474 –
cular complications of diabetes. BMJ. 2006;333:475– 480. 6479.
2. Klein R, Klein BE, Moss SE. Epidemiology of proliferative diabetic 27. Uribarri J, Cai W, Sandu O, Peppa M, Goldberg T, Vlassara H.
retinopathy. Diabetes Care. 1992;15:1875–1891. Diet-derived advanced glycation end products are major contrib-
4872 Stitt IOVS, October 2010, Vol. 51, No. 10

utors to the body’s AGE pool and induce inflammation in healthy 46. Team W. Effect of intensive therapy on the microvascular com-
subjects. Ann N Y Acad Sci. 2005;1043:461– 466. plications of type 1 diabetes mellitus. Jama. 2002;287:2563–
28. Vlassara H, Cai W, Crandall J, et al. Inflammatory mediators are 2569.
induced by dietary glycotoxins, a major risk factor for diabetic 47. Stitt AW. Advanced glycation: an important pathological event in
angiopathy. Proc Natl Acad Sci U S A. 2002;99:15596 –15601. diabetic and age related ocular disease. Br J Ophthalmol. 2001;
29. Kuhla B, Luth HJ, Haferburg D, Boeck K, Arendt T, Munch G. 85:746 –753.
Methylglyoxal, glyoxal, and their detoxification in Alzheimer’s 48. Stitt AW, Moore JE, Sharkey JA, et al. Advanced glycation end
disease. Ann N Y Acad Sci. 2005;1043:211–216. products in vitreous: structural and functional implications for
30. Shinohara M, Thornalley PJ, Giardino I, et al. Overexpression of diabetic vitreopathy. Invest Ophthalmol Visual Sci. 1998;39:
glyoxalase-I in bovine endothelial cells inhibits intracellular ad- 2517–2523.
vanced glycation endproduct formation and prevents hypergly- 49. Fosmark DS, Bragadottir R, Stene-Johansen I, et al. Increased
cemia-induced increases in macromolecular endocytosis. J Clin vitreous levels of hydroimidazolone in type 2 diabetes patients
Invest. 1998;101:1142–1147. are associated with retinopathy: a case-control study. Acta Oph-
31. Yao D, Taguchi T, Matsumura T, et al. High glucose increases thalmol Scand. 2007;85:618 – 622.
angiopoietin-2 transcription in microvascular endothelial cells 50. Gardiner TA, Anderson HR, Stitt AW. Inhibition of advanced
through methylglyoxal modification of mSin3A. J Biol Chem. glycation end-products protects against retinal capillary basement
2007;282:31038 –31045. membrane expansion during long-term diabetes. J Pathol. 2003;
32. Miller AG, Smith DG, Bhat M, Nagaraj RH. Glyoxalase I is critical 201:328 –333.
for human retinal capillary pericyte survival under hyperglycemic 51. Hammes HP, Alt A, Niwa T, et al. Differential accumulation of
conditions. J Biol Chem. 2006;281:11864 –11871. advanced glycation end products in the course of diabetic reti-
nopathy. Diabetologia. 1999;42:728 –736.
33. Morcos M, Du X, Pfisterer F, et al. Glyoxalase-1 prevents mito-
chondrial protein modification and enhances lifespan in Caeno- 52. Murata T, Nagai R, Ishibashi T, Inomuta H, Ikeda K, Horiuchi S.
rhabditis elegans. Aging cell. 2008;7:260 –269. The relationship between accumulation of advanced glycation
end products and expression of vascular endothelial growth
34. Schlotterer A, Kukudov G, Bozorgmehr F, et al. C. elegans as
factor in human diabetic retinas. Diabetologia. 1997;40:764 –
model for the study of high glucose-mediated life span reduction.
769.
Diabetes. 2009;58:2450 –2456.
53. Schalkwijk CG, Ligtvoet N, Twaalfhoven H, et al. Amadori albu-
35. Yu Y, Lyons TJ. A lethal tetrad in diabetes: hyperglycemia, dys-
min in type 1 diabetic patients: correlation with markers of
lipidemia, oxidative stress, and endothelial dysfunction. Am J endothelial function, association with diabetic nephropathy, and
Med Sci. 2005;330:227–232. localization in retinal capillaries. Diabetes. 1999;48:2446 –2453.
36. Onorato JM, Jenkins AJ, Thorpe SR, Baynes JW. Pyridoxamine, an 54. Stitt AW, Li YM, Gardiner TA, Bucala R, Archer DB, Vlassara H.
inhibitor of advanced glycation reactions, also inhibits advanced Advanced glycation end products (AGEs) co-localize with AGE
lipoxidation reactions: mechanism of action of pyridoxamine. receptors in the retinal vasculature of diabetic and of AGE-infused
J Biol Chem. 2000;275:21177–21184. rats. Am J Pathol. 1997;150:523–531.
37. Miyata T, Inagi R, Asahi K, et al. Generation of protein carbonyls 55. Hammes HP, Brownlee M, Edelstein D, Saleck M, Martin S, Fed-
by glycoxidation and lipoxidation reactions with autoxidation erlin K. Aminoguanidine inhibits the development of accelerated
products of ascorbic acid and polyunsaturated fatty acids. FEBS diabetic retinopathy in the spontaneous hypertensive rat. Diabe-
Lett. 1998;437:24 –28. tologia. 1994;37:32–35.
38. Zhang X, Lai Y, McCance DR, et al. Evaluation of N (epsilon)-(3- 56. Karachalias N, Babaei-Jadidi R, Ahmed N, Thornalley PJ. Accumu-
formyl-3,4-dehydropiperidino)lysine as a novel biomarker for the lation of fructosyl-lysine and advanced glycation end products in
severity of diabetic retinopathy. Diabetologia. 2008;51:1723– the kidney, retina and peripheral nerve of streptozotocin-induced
1730. diabetic rats. Biochem Soc Trans. 2003;31:1423–1425.
39. Ono Y, Aoki S, Ohnishi K, Yasuda T, Kawano K, Tsukada Y. 57. Stitt A, Gardiner TA, Alderson NL, et al. The AGE inhibitor
Increased serum levels of advanced glycation end-products and pyridoxamine inhibits development of retinopathy in experimen-
diabetic complications. Diabetes Res Clin Pract. 1998;41:131– tal diabetes. Diabetes. 2002;51:2826 –2832.
137. 58. Agardh E, Bruun A, Agardh CD. Retinal glial cell immunoreactivity
40. Sugiyama S, Miyata T, Ueda Y, et al. Plasma levels of pentosidine and neuronal cell changes in rats with STZ-induced diabetes.
in diabetic patients: an advanced glycation end product. J Am Soc Curr Eye Res. 2001;23:276 –284.
Nephrol. 1998;9:1681–1688. 59. de Gooyer TE, Stevenson KA, Humphries P, Simpson DA,
41. Yamaguchi M, Nakamura N, Nakano K, et al. Immunochemical Gardiner TA, Stitt AW. Retinopathy is reduced during experimen-
quantification of crossline as a fluorescent advanced glycation tal diabetes in a mouse model of outer retinal degeneration.
endproduct in erythrocyte membrane proteins from diabetic pa- Invest Ophthalmol Vis Sci. 2006;47:5561–5568.
tients with or without retinopathy. Diabet Med. 1998;15:458 – 60. Mizutani M, Gerhardinger C, Lorenzi M. Müller cell changes in
462. human diabetic retinopathy. Diabetes. 1998;47:445– 449.
42. Fosmark DS, Torjesen PA, Kilhovd BK, et al. Increased serum 61. Du Y, Miller CM, Kern TS. Hyperglycemia increases mitochon-
levels of the specific advanced glycation end product methylg- drial superoxide in retina and retinal cells. Free Radic Biol Med.
lyoxal-derived hydroimidazolone are associated with retinopathy 2003;35:1491–1499.
in patients with type 2 diabetes mellitus. Metabol Clin Exp. 62. Li Q, Zemel E, Miller B, Perlman I. NADPH diaphorase activity in
2006;55:232–236. the rat retina during the early stages of experimental diabetes.
43. Wagner Z, Wittmann I, Mazak I, et al. N(epsilon)-(carboxymeth- Graefes Arch Clin Exp Ophthalmol. 2003;241:747–756.
yl)lysine levels in patients with type 2 diabetes: role of renal 63. Puro DG. Diabetes-induced dysfunction of retinal Müller cells.
function. Am J Kidney Dis. 2001;38:785–791. Trans Am Ophthalmol Soc. 2002;100:339 –352.
44. Sell DR, Lapolla A, Odetti P, Fogarty J, Monnier VM. Pentosidine 64. Lieth E, Barber AJ, Xu B, et al. Glial reactivity and impaired
formation in skin correlates with severity of complications in glutamate metabolism in short-term experimental diabetic reti-
individuals with long-standing IDDM. Diabetes. 1992;41:1286 – nopathy. Penn State Retina Research Group. Diabetes. 1998;47:
1292. 815– 820.
45. Genuth S, Sun W, Cleary P, et al. Glycation and carboxymethyll- 65. Winkler BS, Starnes CA, Sauer MW, Firouzgan Z, Chen SC. Cul-
ysine levels in skin collagen predict the risk of future 10-year tured retinal neuronal cells and Müller cells both show net pro-
progression of diabetic retinopathy and nephropathy in the dia- duction of lactate. Neurochem Int. 2004;45:311–320.
betes control and complications trial and epidemiology of diabe- 66. Romero FJ, Bosch-Morell F, Romero MJ, et al. Lipid peroxidation
tes interventions and complications participants with type 1 products and antioxidants in human disease. Environ Health
diabetes. Diabetes. 2005;54:3103–3111. Perspect. 1998;106(suppl 5):1229 –1234.
IOVS, October 2010, Vol. 51, No. 10 AGEs and Diabetic Retinopathy 4873

67. Loske C, Neumann A, Cunningham AM, et al. Cytotoxicity of a factor in diabetic retinopathy. Graefes Arch Clin Exp Ophthal-
advanced glycation endproducts is mediated by oxidative stress. mol. 2003;241:56 – 62.
J Neural Transm. 1998;105:1005–1015. 89. Brownlee M, Vlassara H, Kooney A, Ulrich P, Cerami A. Amino-
68. Taniguchi N, Kaneto H, Asahi M, et al. Involvement of glycation guanidine prevents diabetes-induced arterial wall protein cross-
and oxidative stress in diabetic macroangiopathy. Diabetes. 1996; linking. Science. 1986;232:1629 –1632.
45(suppl 3):S81–S83. 90. Vasan S, Foiles P, Founds H. Therapeutic potential of breakers of
69. Mokini Z, Marcovecchio ML, Chiarelli F. Molecular pathology of advanced glycation end product-protein crosslinks. Arch Bio-
oxidative stress in diabetic angiopathy: role of mitochondrial and chem Biophys. 2003;419:89 –96.
cellular pathways. Diabetes Res Clin Pract. 2010;87:313–321. 91. Hammes HP, Martin S, Federlin K, Geisen K, Brownlee M. Ami-
70. Zheng L, Kern TS. Role of nitric oxide, superoxide, peroxynitrite noguanidine treatment inhibits the development of experimental
and PARP in diabetic retinopathy. Front Biosci. 2009;14:3974 – diabetic retinopathy. Proc Natl Acad Sci U S A. 1991;88:11555–
3987. 11558.
71. Kowluru RA. Diabetic retinopathy: mitochondrial dysfunction 92. Kern TS, Engerman RL. Pharmacological inhibition of diabetic
and retinal capillary cell death. Antioxid Redox Signal. 2005;7: retinopathy: aminoguanidine and aspirin. Diabetes. 2001;50:
1581–1587. 1636 –1642.
72. Kowluru RA, Koppolu P, Chakrabarti S, Chen S. Diabetes-induced 93. Agardh E, Hultberg B, Agardh C. Effects of inhibition of glycation
activation of nuclear transcriptional factor in the retina, and its and oxidative stress on the development of cataract and retinal
inhibition by antioxidants. Free Radic Res. 2003;37:1169 –1180. vessel abnormalities in diabetic rats. Curr Eye Res. 2000;21:543–
73. Kowluru RA, Engerman RL, Case GL, Kern TS. Retinal glutamate 549.
in diabetes and effect of antioxidants. Neurochem Int. 2001;38: 94. Bolton WK, Cattran DC, Williams ME, et al. Randomized trial of an
385–390. inhibitor of formation of advanced glycation end products in
diabetic nephropathy. Am J Nephrol. 2004;24:32– 40.
74. Kowluru RA, Odenbach S. Effect of long-term administration of
alpha-lipoic acid on retinal capillary cell death and the develop- 95. Ren XY, Li YN, Qi JS, Niu T. Peroxynitrite-induced protein nitra-
ment of retinopathy in diabetic rats. Diabetes. 2004;53:3233– tion contributes to liver mitochondrial damage in diabetic rats. J
3238. Diabetes Complications. 2008;22:357–364.
75. Kowluru RA, Kowluru V, Xiong Y, Ho YS. Overexpression of 96. Khalifah RG, Baynes JW, Hudson BG. Amadorins: novel post-
mitochondrial superoxide dismutase in mice protects the retina Amadori inhibitors of advanced glycation reactions. Biochem
from diabetes-induced oxidative stress. Free Radic Biol Med. Biophys Res Commun. 1999;257:251–258.
2006;41:1191–1196. 97. Voziyan PA, Metz TO, Baynes JW, Hudson BG. A post-Amadori
inhibitor pyridoxamine also inhibits chemical modification of
76. Gardiner TA, Archer DB, Curtis TM, Stitt AW. Arteriolar involve-
proteins by scavenging carbonyl intermediates of carbohydrate
ment in the microvascular lesions of diabetic retinopathy: impli-
and lipid degradation. J Biol Chem. 2002;277:3397–3403.
cations for pathogenesis. Microcirculation. 2007;14:25–38.
98. Rahbar S. Novel inhibitors of glycation and AGE formation. Cell
77. Li W, Yanoff M, Jian B, He Z. Altered mRNA levels of antioxidant
Biochem Biophys. 2007;48:147–157.
enzymes in pre-apoptotic pericytes from human diabetic retinas.
Cell Mol Biol (Noisy-le-grand). 1999;45:59 – 66. 99. Bhatwadekar A, Glenn JV, Figarola JL, et al. A new advanced
glycation inhibitor, LR-90, prevents experimental diabetic reti-
78. Chen BH, Jiang DY, Tang LS. Advanced glycation end-products
nopathy in rats. Br J Ophthalmol. 2008;92:545–547.
induce apoptosis involving the signaling pathways of oxidative
100. Gerhardinger C, Dagher Z, Sebastiani P, Seek Park Y, Lorenzi M.
stress in bovine retinal pericytes. Life Sci. 2006;79:1040 –1048.
The transforming growth factor-beta pathway is a common target
79. Yamagishi S, Fujimori H, Yonekura H, Tanaka N, Yamamoto H. of drugs that prevent experimental diabetic retinopathy. Diabe-
Advanced glycation endproducts accelerate calcification in mi- tes. 2009;58:1659 –1667.
crovascular pericytes. Biochem Biophys Res Commun. 1999;
101. Brucklacher RM, Patel KM, VanGuilder HD, et al. Whole genome
258:353–357.
assessment of the retinal response to diabetes reveals a progres-
80. Hughes SJ, Wall N, Scholfield CN, et al. Advanced glycation sive neurovascular inflammatory response. BMC Med Genomics.
endproduct modified basement membrane attenuates endothe- 2008;1:26.
lin-1 induced [Ca2⫹]i signalling and contraction in retinal micro- 102. Vincent JA, Mohr S. Inhibition of caspase-1/interleukin-1beta sig-
vascular pericytes. Mol Vis. 2004;10:996 –1004. naling prevents degeneration of retinal capillaries in diabetes and
81. Liu B, Bhat M, Padival AK, Smith DG, Nagaraj RH. Effect of galactosemia. Diabetes. 2007;56:224 –230.
dicarbonyl modification of fibronectin on retinal capillary peri- 103. Chan PS, Kanwar M, Kowluru RA. Resistance of retinal inflamma-
cytes. Invest Ophthalmol Visual Sci. 2004;45:1983–1995. tory mediators to suppress after reinstitution of good glycemic
82. Mamputu JC, Renier G. Signalling pathways involved in retinal control: novel mechanism for metabolic memory. J Diabetes
endothelial cell proliferation induced by advanced glycation end Complications. 2010;24:55– 63.
products: inhibitory effect of gliclazide. Diabetes Obes Metab. 104. Joussen AM, Poulaki V, Le ML, et al. A central role for inflamma-
2004;6:95–103. tion in the pathogenesis of diabetic retinopathy. FASEB J. 2004;
83. Chibber R, Molinatti PA, Rosatto N, Lambourne B, Kohner EM. 18:1450 –1452.
Toxic action of advanced glycation end products on cultured 105. Kuiper EJ, Witmer AN, Klaassen I, Oliver N, Goldschmeding R,
retinal capillary pericytes and endothelial cells: relevance to dia- Schlingemann RO. Differential expression of connective tissue
betic retinopathy. Diabetologia. 1997;40:156 –164. growth factor in microglia and pericytes in the human diabetic
84. Stitt AW, McGoldrick C, Rice-McCaldin A, et al. Impaired retinal retina. Br J Ophthalmol. 2004;88:1082–1087.
angiogenesis in diabetes: role of advanced glycation end products 106. Zeng HY, Green WR, Tso MO. Microglial activation in human
and galectin-3. Diabetes. 2005;54:785–794. diabetic retinopathy. Arch Ophthalmol. 2008;126:227–232.
85. Padayatti PS, Jiang C, Glomb MA, Uchida K, Nagaraj RH. High 107. Rungger-Brandle E, Dosso AA, Leuenberger PM. Glial reactivity,
concentrations of glucose induce synthesis of argpyrimidine in an early feature of diabetic retinopathy. Invest Ophthalmol Vi-
retinal endothelial cells. Curr Eye Res. 2001;23:106 –115. sual Sci. 2000;41:1971–1980.
86. Moore TC, Moore JE, Kaji Y, et al. The role of advanced glycation 108. Zeng XX, Ng YK, Ling EA. Neuronal and microglial response in
end products in retinal microvascular leukostasis. Invest Ophthal- the retina of streptozotocin-induced diabetic rats. Vis Neurosci.
mol Visual Sci. 2003;44:4457– 4464. 2000;17:463– 471.
87. Canning P, Glenn JV, Hsu DK, Liu FT, Gardiner TA, Stitt AW. 109. Kislinger T, Fu C, Huber B, et al. N(epsilon)-(carboxymethyl)l-
Inhibition of advanced glycation and absence of galectin-3 pre- ysine adducts of proteins are ligands for receptor for advanced
vent blood-retinal barrier dysfunction during short-term diabetes. glycation end products that activate cell signaling pathways and
Exp Diabetes Res. 2007;2007:51837. modulate gene expression. J Biol Chem. 1999;274:31740 –31749.
88. Xu X, Li Z, Luo D, et al. Exogenous advanced glycosylation end 110. Liliensiek B, Weigand MA, Bierhaus A, et al. Receptor for ad-
products induce diabetes-like vascular dysfunction in normal rats: vanced glycation end products (RAGE) regulates sepsis but not
4874 Stitt IOVS, October 2010, Vol. 51, No. 10

the adaptive immune response. J Clin Invest. 2004;113:1641– complications, neurodegeneration, and inflammation. Ann Med.
1650. 2009;25:1–15.
111. Hofmann MA, Drury S, Fu C, et al. RAGE mediates a novel 122. Soulis T, Thallas V, Youssef S, et al. Advanced glycation end
proinflammatory axis: a central cell surface receptor for S100/ products and their receptors co-localise in rat organs susceptible
calgranulin polypeptides. Cell. 1999;97:889 –901. to diabetic microvascular injury. Diabetologia. 1997;40:619 –
112. Yan SD, Chen X, Fu J, et al. RAGE and amyloid-beta peptide 628.
neurotoxicity in Alzheimer’s disease. Nature. 1996;382:685– 691. 123. Pachydaki SI, Tari SR, Lee SE, et al. Upregulation of RAGE and its
113. Brett J, Schmidt AM, Yan SD, et al. Survey of the distribution of a ligands in proliferative retinal disease. Exp Eye Res. 2006;82:807–
newly characterized receptor for advanced glycation end prod- 815.
ucts in tissues. Am J Pathol. 1993;143:1699 –1712. 124. Howes KA, Liu Y, Dunaief JL, et al. Receptor for advanced
114. Chavakis T, Bierhaus A, Al-Fakhri N, et al. The pattern recognition glycation end products and age-related macular degeneration.
receptor (RAGE) is a counterreceptor for leukocyte integrins: a Invest Ophthalmol Visual Sci. 2004;45:3713–3720.
novel pathway for inflammatory cell recruitment. J Exp Med. 125. Barile GR, Pachydaki SI, Tari SR, et al. The RAGE axis in early
2003;198:1507–1515. diabetic retinopathy. Invest Ophthalmol Visual Sci. 2005;46:
2916 –2924.
115. Schmidt AM, Stern DM. RAGE: a new target for the prevention
and treatment of the vascular and inflammatory complications of 126. Yao D, Brownlee M. Hyperglycemia-induced reactive oxygen
diabetes. Trends Endocrinol Metab. 2000;11:368 –375. species increase expression of the receptor for advanced glyca-
tion end products (RAGE) and RAGE ligands. Diabetes. 2010;59:
116. Bierhaus A, Humpert PM, Morcos M, et al. Understanding RAGE,
249 –255.
the receptor for advanced glycation end products. J Mol Med.
127. Zong H, Ward M, Madden A, Yong PH, Limb GA, Stitt AW.
2005;83:876 – 886.
Hyperglycaemia-induced pro-inflammatory responses by retinal
117. Zong H, Madden A, Ward M, Mooney MH, Elliott CT, Stitt AW. Müller glia responses are regulated by the receptor for advanced
Homodimerisation is essential for the receptor for advanced gly- glycation end-products (RAGE). Diabetologia. 2010. In press.
cation end-products (RAGE)-mediated signal transduction. J Biol
128. Marx N, Walcher D, Ivanova N, et al. Thiazolidinediones reduce
Chem. 2010;23:23137–23146.
endothelial expression of receptors for advanced glycation end
118. Hudson BI, Kalea AZ, Del Mar Arriero M, et al. Interaction of the products. Diabetes. 2004;53:2662–2668.
RAGE cytoplasmic domain with diaphanous-1 is required for 129. Yamagishi S, Takeuchi M. Nifedipine inhibits gene expression of
ligand-stimulated cellular migration through activation of Rac1 receptor for advanced glycation end products (RAGE) in endo-
and Cdc42. J Biol Chem. 2008;283:34457–34468. thelial cells by suppressing reactive oxygen species generation.
119. Hermani A, De Servi B, Medunjanin S, Tessier PA, Mayer D. Drugs Exp Clin Res. 2004;30:169 –175.
S100A8 and S100A9 activate MAP kinase and NF-kappaB signaling 130. Yamagishi S, Takeuchi M, Matsui T, Nakamura K, Imaizumi T,
pathways and trigger translocation of RAGE in human prostate Inoue H. Angiotensin II augments advanced glycation end prod-
cancer cells. Exp Cell Res. 2006;312:184 –197. uct-induced pericyte apoptosis through RAGE overexpression.
120. Chuong C, Katz J, Pauley KM, Bulosan M, Cha S. RAGE expression FEBS Lett. 2005;579:4265– 4270.
and NF-kappaB activation attenuated by extracellular domain of 131. Nakamura K, Yamagishi S, Nakamura Y, et al. Telmisartan inhibits
RAGE in human salivary gland cell line. J Cell Physiol. 2009;221: expression of a receptor for advanced glycation end products
430 – 434. (RAGE) in angiotensin-II-exposed endothelial cells and decreases
121. Yan SF, Du Yan S, Ramasamy R, Schmidt AM. Tempering the serum levels of soluble RAGE in patients with essential hyperten-
wrath of RAGE: An emerging therapeutic strategy against diabetic sion. Microvasc Res. 2005;70:137–141.

You might also like