You are on page 1of 10

Diabetes & Metabolism 36 (2010) 1–10

Review

Advanced glycation end-products: Implications for diabetic


and non-diabetic nephropathies
M. Daroux a,b , G. Prévost a,c , H. Maillard-Lefebvre a,d , C. Gaxatte a,e ,
V.D. D’Agati f , A.M. Schmidt g , É. Boulanger a,e,∗
a EA2693, Vascular Aging Biology, Research Division, Lille Medical School, 1, place de Verdun, 59045 Lille, France
b Department of Nephrology, Regional University Hospital of Lille, Lille, France
c Department of Endocrinology and Diabetology, Regional University Hospital of Lille, Lille, France
d Department of Internal Medicine, Regional University Hospital of Lille, Lille, France
e Department of Gerontology, Regional University Hospital of Lille, Lille, France
f Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
g Department of Surgical Science, College of Physicians and Surgeons, Columbia University, New York, NY, USA

Received 10 January 2009; accepted 23 June 2009


Available online 22 November 2009

Abstract
Glycation is the process whereby sugars bind to the free amine residues of proteins. These newly formed modified molecular species are known
as ‘advanced glycation end-products’, or AGEs. AGE toxicity may occur through at least three mechanisms: interaction with the receptor for AGEs
(RAGE); tissue deposition; and in situ glycation. AGEs trigger proinflammatory, profibrotic and procoagulant cellular responses that are capable
of damaging tissues, often targeting particular organs. In diabetic patients, the conditions needed to promote AGE formation are all present, and
are further accentuated by accompanying renal failure. The aim of this review is to outline the involvement of AGEs in the various forms of renal
pathology associated with diabetic and non-diabetic nephropathies. AGEs are present in all renal compartments in diabetic patients, including the
vessels, glomeruli, tubules and interstitium. Many cell types may be activated—specifically, endothelial, tubular and mesangial cells, and podocytes.
AGEs play a major role in the accumulation of extracellular matrix, as occurs in diabetic glomerulosclerosis, and are also involved in most diabetic
(renovascular, microangiopathic and glomerular) and non-diabetic renal injury associated with progressive glomerulosclerosis and ageing.
© 2009 Elsevier Masson SAS. All rights reserved.

Keywords: Advanced glycation end-products; Diabetic nephropathy; Glomerulosclerosis; Fibrosis; RAGE; Review

Résumé
Produits de la glycation avancée : implication au cours de la néphropathie diabétique et des autres néphropathies.
La glycation est une réaction non enzymatique définie par la fixation d’un sucre à un résidu amine libre d’une protéine. Ces molécules néoformées
ont des effets délétères et constituent une nouvelle famille nommée « produits de glycation avancée » ou AGE (advanced glycation end-products).
La toxicité des AGE est fondée sur trois mécanismes : l’interaction avec le récepteur des AGE (RAGE), les dépôts tissulaires et la glycation in
situ. Les AGE entraînent une réponse cellulaire néfaste, pro-inflammatoire, profibrosante et procoagulante, tant au niveau tissulaire qu’au niveau
de certains organes cibles. Au cours du diabète, toutes les conditions sont réunies pour promouvoir la formation des AGE, notamment en cas
d’insuffisance rénale associée. L’objectif de cette revue est de décrire l’implication des AGE dans les diverses pathologies rénales associées ou
non au diabète. Les AGE sont présents, chez le diabétique, dans toutes les structures rénales incluant les vaisseaux, les glomérules, les tubules et
l’interstitium. De nombreux types cellulaires peuvent ainsi être activés : les cellules endothéliales, tubulaires, mésangiales ainsi que les podocytes.
Les AGE jouent également un rôle majeur dans l’accumulation de la matrice extracellulaire mise en évidence au cours du diabète et participent aux
autres complications rénales liées au diabète (néphroangiosclérose et glomérulopathie). Les AGE sont également impliqués dans les néphropathies
non diabétiques et au cours du vieillissement rénal.
© 2009 Elsevier Masson SAS. Tous droits réservés.

Mots clés : Produits de glycation avancée ; Néphropathie diabétique ; Glomérulosclerose ; Fibrose ; RAGE ; Revue générale

∗ Corresponding author. Tel.: +33 0 6 62 70 05 67; fax: +33 0 3 20 44 64 87.


E-mail address: eric.boulanger@chru-lille.fr (É. Boulanger).

1262-3636/$ – see front matter © 2009 Elsevier Masson SAS. All rights reserved.
doi:10.1016/j.diabet.2009.06.005
2 M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10

1. Introduction

Diabetes-induced renal disorders can affect multiple com-


partments of the kidney, as implied by the term ‘diabetic
nephropathies’, including: the large vessels, with the subse-
quent development of atherosclerosis and renovascular disease;
the microvessels, in the form of arterio- and arteriolosclerosis;
and, in particular, the glomeruli, leading to nodular or dif-
fuse glomerulosclerosis, generally considered the quintessential
lesion of diabetic nephropathy (DN) [1]. These pathological
changes arise in part from the adverse effects of hyperglycaemia,
leading to the formation of advanced glycation end-products
(AGEs) [2,3]. AGEs are also implicated in the pathophysiol-
ogy of non-diabetic renal diseases and ageing. They have been
found in renal tissues in IgA and lupus nephritis, hyperten-
sive nephropathy, renal fibrosis and chronic transplant rejection
[4–6], and are also implicated in physiological ageing as well as Fig. 1. Diagrammatic representation of advanced glycation end-product (AGE)
age-related disorders such as Alzheimer’s disease and macular formation.
degeneration [7]. Diabetologists, along with nephrologists and
geriatricians, need to be familiar with the deleterious effects of
widely studied, as they are antigenic and easy to analyze:
AGE formation and resulting organ damage. The present review
N␧ -(carboxylmethyl)lysine (CML), which is mainly present in
is an update of recent advances in our understanding of the roles
biological fluids and has a short half-life; and pentosidine, which
of AGEs in the development of DN, non-diabetic renal disease
is mostly found in the extracellular matrix (ECM) and, thus, has
and renal ageing.
a longer half-life. AGE toxicity is linked to three mechanisms:
deposition; in situ glycation and receptor interaction.
2. AGEs and their interaction with receptors

Chronic hyperglycaemia promotes the formation of modified 2.1. AGE receptors


molecular species such as AGEs. AGEs result from the binding
of free lysine or arginine NH2 residues on proteins to a sugar with At least five types of AGE receptors have been identified
an ‘ose’, such as glucose, fructose or glucose-6-phosphate. The so far: the macrophage scavenger receptor 1 (MSR1 or CD36)
formation of AGEs can occur through three major biochemical [9]; the AGE receptor R1 (AGE-R1 or p60), corresponding to
mechanisms: glycation; the polyol pathway and glycoxidation. oligosaccharyl transferase 48; the AGE receptor R2 (AGE-R2
The first, glycation, directly depends on glucose concentra- or p90), which corresponds to phosphoprotein 80K-H; the AGE
tions, time and temperature, and requires several stages before receptor R3 (AGE-R3 or galactin-3), a scavenger receptor that
an AGE can be formed. First described in 1912 by Louis-Camille recognizes galactoside residues; and RAGE, commonly referred
Maillard at the Paris Academy of Sciences, glycation is a non- to as the ‘receptor for AGEs’ and the most widely studied type
enzymatic process in which glucose binds to a protein in a [10].
high-glucose-concentration environment [8]. Glycation of pro- RAGE, a transmembrane receptor of the immunoglobulin
teins involves the formation of a Schiff base, which is then superfamily [11], is a multiligand receptor with the capability
transformed into Amadori products that, in turn, are subject of interacting with several types of proteins, including AGEs,
to further molecular modifications before becoming an AGE. the S100 proteins or calgranulins (belonging to the proinflam-
The most familiar intermediary glycation product measured in matory cytokines), the high-mobility group B1 (HMGB1) also
routine clinical practice is glycated haemoglobin (HbA1c ). known as ‘amphoterin’, and the proteins with a ␤ fibrillary
The second mechanism, the polyol pathway, is enzymatic in structure [12,13]. In humans, the RAGE gene is located on chro-
nature. Glucose is transformed by aldolase reductase or sorbitol mosome 6 in the major histocompatibility complex (MHC) class
dehydrogenase into an intermediary product that, after binding III region [14]. RAGE consists of an intracellular domain, a short
to a protein, becomes an AGE. transmembrane domain and an extracellular domain. Genetic
The glycoxidation pathway is based on oxidative stress, variants of the RAGE gene (AGER in HUGO nomenclature)
whereby the oxidation of glucose leads to the formation of gly- have been associated with vascular disease and risk of renal
oxal and methylglyoxal. These oxidized sugars are extremely disorders [15]. The association between the RAGE -374 T/A
unstable and can quickly react with different proteins to form homozygous AA genotype and cardiovascular disease as well as
AGEs (Fig. 1). albumin excretion in type 1 diabetic patients with poor metabolic
Although there is no precise AGE classification, these control suggests a gene–environment interaction in the develop-
molecules can be differentiated according to the type of ment of DN and cardiovascular complications [16]. The 82 Ser
binding, the nature of the glycated protein or the particular allele of the RAGE gene is a risk allele for developing advanced
sugar that is bound. So far, two AGEs have been especially nephropathy in Caucasian type 1 diabetic patients [17].
M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10 3

tease (MMP) inhibitors. Cells deficient in ADAM10 produce


only a small amount of sRAGE, suggesting that it is responsible
for RAGE cleavage. Both esRAGE and cRAGE are functionally
equivalent and able to interact with RAGE ligands.
RAGE is particularly found in the vascular compartment
(endothelial cells, vascular smooth muscle cells, mononuclear
leukocytes, macrophages), the nervous system, and in the lungs,
muscles and peritoneum [20–22]. In the kidneys, RAGE has
been identified on the surface of endothelial cells, podocytes,
tubular cells and mesangial cells [23,24].

2.2. Interaction of AGE and RAGE

The AGE/RAGE interaction activates a series of intra-


cellular signalling pathways, including nicotinamide adenine
dinucleotide phosphate (NADPH) oxidase and nuclear fac-
tor kappa-B (NF␬-B), with production of reactive oxygen
species (ROS) [25]. The cellular response can involve sev-
eral types—proinflammatory, profibrotic, procoagulant and/or
angiogenic—with overexpression of cell adhesion molecules
such as vascular cell adhesion molecule-1 (VCAM-1), and the
production of cytokines [interleukin-2 (IL-2), IL-6 and tumour
necrosis factor-␣ (TNF-␣)], tissue factor (TF) and vascular
endothelial growth factor (VEGF; Fig. 2B) [22,26–28].

3. AGEs in diabetic nephropathies

3.1. Where are AGEs located in the diabetic kidney?

AGEs can be identified in many renal structures. In arteries,


AGE deposits are mainly found in atheromatous plaques. Their
presence has been reported in the carotid, aortic, femoral, radial
and iliac arteries. However, there are no reports in the literature
of the presence of AGEs in the renal artery, although such a
finding appears to be reasonable [29,30]. The AGE concentration
in plaques is correlated with the degree of atherosclerosis, and
is even higher in diabetic patients with renal failure [25,31]. An
Fig. 2. A. Soluble RAGE (sRAGE) cleavage and splicing: RAGE: recep-
ultrastructural study carried out in rats with DN demonstrated
tor of advanced glycation end-products (AGEs); cRAGE: cleaved RAGE; the presence of AGEs in the glomerular basement membrane,
esRAGE: endogenous secretory RAGE. B. Cell response after RAGE activation: mesangium, podocytes, tubules and endothelial cells [23,32].
ICAM-1: intercellular adhesion molecule-1; VCAM-1: vascular cellular adhe- The extent of AGE concentration is proportional to the severity
sion molecule-1; IL-6: interleukin-6, TNF-␣: tumour necrosis factor-␣; NFk B: of mesangial expansion in the course of DN [33] (Table 1).
nuclear factor-kappa B.

3.2. Cellular effects of AGEs


Soluble RAGE (sRAGE) corresponds to the extracellu-
lar domain of RAGE. As the N-terminal ‘V’-type domain Endothelial cells express RAGE, where interaction with
is also included, sRAGE has the same ligand-binding speci- AGEs can mediate numerous negative effects leading to macro-
ficity as RAGE and may act as a decoy by binding with and microvascular endothelial dysfunction. Both in vitro and in
proinflammatory ligands and preventing them from reaching vivo, endothelial RAGE activation leads to increased expres-
membrane-associated RAGE. sRAGE comprises two forms: sion of VCAM-1 (blood cell–endothelium interactions) and
endogenous secretory RAGE (esRAGE) and cleaved RAGE E-cadherin (homotype adhesion) [34–36]. The glycation of
(cRAGE; Fig. 2A) [18,19]. The former occurs via alternative endothelial intracellular proteins has also been described. In
splicing of intron 9/exon 10 of the RAGE gene [18]. Another vitro, glycation of fibroblast growth factor-␤ (FGF-␤) is accom-
mechanism responsible for the circulating pool of sRAGE is panied by a significant decrease in binding of heparin, its natural
based on proteolytic processing (by ADAM10) from the full- ligand, and by a reduction in its mitogenic activity [37]. AGEs
length membrane-bound RAGE that produces cRAGE [19]. The directly lower the level of nitric oxide synthase (NOS) expres-
formation of cRAGE can be inhibited with matrix metallopro- sion [38]. They can also directly interact with NO and inactivate
4 M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10

Table 1
AGE deposits in diabetic kidneys, as reflected by the percentage of AGE-positive staining in type 2 diabetic renal biopsies.
Mesangium Glomerular basement membrane Tubular basement membrane Interstitium Vessel walls

HSA-CML (%) 96 4 85 19 96
Pentosidine (%) 77 4 31 90 54

Data are based on a study of 26 human PBR [27]. HSA-CML: human serum albumin-carboxymethyl-lysine.

it [39], and disrupt endothelial anticoagulant activities by reduc- the presence of mesenchymal markers such as ␣-smooth muscle
ing levels of the anticoagulant thrombomodulin and increasing actin, and plays a key role in the development of renal fibrosis
levels of procoagulant TF [40]. Endothelial RAGE activation by stimulating the secretion of ECM proteins such as collagen
by AGEs stimulates the synthesis of ROS through activation of and fibronectin [3]. In vitro, tubular epithelial cell RAGE acti-
the NADPH-oxidase pathway, with activation of superoxide ion vation by AGEs induces EMT via the TGF-␤ pathway, whereas
(O2 – ) and hydrogen peroxide (H2 O2 ). These factors all promote EMT may be totally inhibited by anti-RAGE antibodies and par-
endothelial dysfunction through perturbations of coagulation, tially inhibited by anti-TGF-␤ antibodies [56]. In diabetic rats,
permeability, vasomotor function and cell adhesion, leading to 0.5–5% of tubular cells express myofibroblast markers. Renal
the development of renal macro- and microvascular diabetic biopsies from patients with DN also demonstrate the presence
lesions [41,42]. of EMT [56].
Mesangial cell RAGE activation has effects on the cell cycle
and maintains mesangial cells in a quiescent state. In turn, this 3.3. Effects of AGEs on ECM
inhibition of cell proliferation promotes mesangial cell apoptosis
and hypertrophy [43,44]. AGEs increase mesangial synthesis of Glycated proteins can modify ECM structure by cross-linking
fibronectin as well as collagen types I and IV. Following RAGE proteins such as collagen and elastin. This process increases
activation, mesangial cells secrete monocyte chemoattractant ECM rigidity, reduces its turnover and might, at least partly,
protein-1 (MCP-1), which participates in the inflammatory pro- explain the vascular rigidity and hypertension that typically
cess [44,45]. Mesangial cells also express other receptors for develop during diabetes and ageing [57].
AGE, such as AGE-R1 and AGE-R2, the activation of which DN progression is accompanied by significant mesangial
may contribute to the damaging effects of AGEs by promoting expansion that is partly linked to a decrease in ECM degra-
mesangial cell apoptosis [46,47]. dation and reversible in the presence of aminoguanidine, an
Podocyte RAGE activation is strongly implicated in the inhibitor of AGE formation [58]. Modification in matrix turnover
development of DN [48,49]. AGEs interfere with the podocyte includes regulation of MMP, and of their activators [membrane-
cell cycle and inhibit cell proliferation, causing cytoplasmic type MMP (MT-MMP)] and inhibitors [tissue inhibitor MMP
hypertrophy followed by apoptosis. This effect can be pre- (TIMP)]. The collagen affinity for MMP-3 is clearly diminished
vented by RAGE small interfering RNA (siRNA) [50]. Glycated when the latter is glycated [59]. While MMP-2 gene expression
albumin, through the engagement of RAGE, inhibits nephrin augments this activity, the presence of AGEs in the mesangium
synthesis by podocytes, and alters the slit diaphragm that forms reduces the activity of MT1-MMP (an MMP-2 activator) by
the glomerular filtration barrier [51]. AGEs also interact with 45%. Also, there is a parallel augmentation of the activity of
podocyte cytoskeletal protein ␣-actinin-4 (ACTN-4), a key com- inhibitors TIMP-1 and TIMP-2, although these effects can be
ponent of the slit diaphragm [52]. In the presence of AGEs, prevented by aminoguanidine, further emphasizing the impor-
ACTN-4 gene transcription and expression by podocytes is sig- tant role of AGEs in disease progression [60]. MMP activity in
nificantly diminished [53]. This alteration is an initial step in the non-collagen proteins such as MMP-7 is also lowered in the pres-
development of proteinuria, and diminution of ACTN-4 expres- ence of AGEs [61] (Fig. 3). ECM glycation leads to alterations in
sion has been correlated with the degree of albuminuria and renal the cell–matrix interaction. In vitro, the culture of podocytes on
failure [54]. a glycated matrix is accompanied by a reduction in cell attach-
AGE engagement with RAGE on tubular cells leads to ment and proliferation [62]. AGEs also regulate ECM protein
TGF-␤ pathway activation. In vitro, RAGE activation by cul- and protease expression by human mesangial cells [8].
tured epithelial tubular cells is followed by an increase of
TGF-␤ concentration in the culture medium, and by enhance- 3.4. From animal models to humans
ment of transcription and expression of small mothers against
decapentaplegic-2 and -3 (SMAD-2 and SMAD-3). These two The presence of AGEs in the kidney is the result of different
molecules, which are activated early in the process, affect mechanisms: trapping; in situ glycation and tubular reabsorp-
the transcription of numerous genes [55]. TGF-␤ induces the tion. Compared with control animals, intravenous injection
epithelial-to-mesenchymal transition (EMT) process, which of AGEs over a 5-month period in normal rats led to a 50%
involves phenotype modification of epithelial tubular cells into increase in renal accumulation of AGEs [63]. AGE distribution
myofibroblastic cells, a transformation that is accompanied by was comparable to that observed in DN and mimicked the
altered cell function favouring profibrotic activity. EMT results histological changes of DN, including hypertrophy and sclerosis
in phenotype changes, with a decrease in epithelial markers and of glomeruli, mesangial matrix hypertrophy, thickening of
M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10 5

4. AGEs and diabetic nephropathy: similarities with


other nephropathies and ageing

DN is not the only kidney condition involving AGEs. Renal


disorders linked with AGEs are also reported during CRF,
chronic inflammatory diseases, high dietary AGE and ageing,
although the precise role of AGEs in these different kidney
pathologies has yet to be elucidated. During diabetes, AGEs
are a consequence of chronic hyperglycaemia and precede dia-
betic complications [70]. In non-diabetic patients, their presence
could disrupt the equilibrium between dietary intake, oxidation
(glycoxidation), in situ glycation and renal elimination. Exces-
sive kidney AGE deposition is found in arterionephrosclerosis,
IgA nephropathy, lupus nephritis, uraemia and the experimental
murine model of focal sclerosis mediated by adriamycin [23,71].
Compared with DN, AGEs and their distribution often differ in
these diverse pathological settings (Table 2).
All biopsies preformed in patients with arterionephrosclero-
sis have been strongly positive for CML levels in the glomeruli
and vascular structures, with pentosidine being mostly present
Fig. 3. Double effects of advanced glycation end-products (AGEs) on matrix
in the interstitium [23]. AGEs are known to be important in
metalloproteinases (MMPs). MT-MMPs: membrane-type MMPs; TIMPs: tissue
inhibitors of MMPs; ECM: extracellular matrix. atherosclerosis progression and are positively correlated with
cardiovascular mortality in non-diabetic women [72]. Dur-
ing IgA nephropathy, CML and pentosidine staining in the
tubular and glomerular basement membranes, and effacement of mesangium and interstitium is particularly mild, whereas these
podocyte foot processes. AGE renal accumulation was accom- two types of AGEs are found in excess in the tubules. Also, there
panied by increased excretion of both total urinary protein (2.5 are high levels of pyrraline in the interstitium, while vascular
times that of the controls) and urinary AGE. Injection of AGEs staining for AGEs is faint [33,73]. In lupus nephritis, patho-
into non-diabetic rats that had undergone subtotal nephrectomy logical lesions are present in the glomeruli and, in some cases,
led to glomerulosclerosis and tubulointerstitial damage [63,64]. the vessels, too, with a similar distribution, but weaker intensity
A study of rats given radioactive pentosidine confirmed the of staining of AGEs compared with DN. The increased AGEs
key role of the kidney in AGE metabolism, with 83% of the found in lupus glomerulonephritis could be the consequence of
total radioactivity eliminated via the urine, and a large residual chronic inflammation that leads to oxidative stress and glycox-
quantity of AGEs trapped in the kidneys—35% compared with idation [74]. In vitro, RAGE activation of mesangial cells by
only 2% in the liver [65]. Urinary elimination of AGE is directly DNA antibody induces a proinflammatory response and may
dependent on dietary intake. Indeed, a reduced AGE intake was contribute to renal changes [75].
followed by lower circulating and tissue AGE levels in mice. In It is uncertain to what extent kidney AGE accumulation is
healthy volunteers, a low-AGE diet was followed by a decrease a cause or consequence of kidney disease, and AGEs could
in pyrraline urinary concentration [66]. These findings have also even exert effects through a sort of vicious circle. During CRF,
been reported in patients with chronic renal failure (CRF) and uraemia itself promotes blood and tissue AGE accumulation.
diabetes [67,68]. In addition, tubular reabsorption plays a key Reduction of AGE clearance and permanent oxidative (car-
role in renal AGE accumulation. Epithelial cells in the proximal bonyl) stress are responsible for such AGE excess [65]. Also, in
tubules express an apical membrane receptor called ‘megalin’, CRF, the presence of AGEs increases endothelial dysfunction
which can reabsorb filtered low-molecular-weight (LMW) associated with an imbalance between relaxation and vasocon-
proteins, including LMW AGEs. After megalin-binding, AGEs striction: endothelial reactivity is decreased, and the response to
undergo endocytosis, and are then directed towards lysosomes ischaemia and hyperthermia is not adaptative [76]. In 51 non-
through a mechanism that is both limited and saturable [65,69]. diabetic uraemic patients, blood levels of AGEs and mRNA of

Table 2
Localization of advanced glycation end-products (AGEs) in renal biopsies from diabetic and non-diabetic nephropathy patients.
Mesangium Glomerular basement membrane Tubular basement membrane Interstitium Vessel walls

Diabetic nephropathy (n = 26) CML CML CML Pentosidine CML


Focal segmental glomerulosclerosis (n = 18) Pentosidine CML Pentosidine Pentosidine CML
Hypertensive nephropathy (n = 7) CML CML CML/Pentosidine Pentosidine CML
IgA nephropathy (n = 15) MDA-lysine ND Pyrraline Pyrraline MDA-lysine
Lupus nephritis (n = 11) CML CML Pentosidine Pentosidine CML

CML: carboxymethyl-lysine; MDA-lysine: malondialdehyde-lysine; ND: no determination; main localizations are shown in bold [27,33,63].
6 M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10

RAGE correlated with the glomerular filtration rate and endothe- retinopathy and neuropathy, and improves endothelial dys-
lial dysfunction. AGEs accelerate degradation of renal function function [91,92]. ACTION 1 (Aminoguanidine Clinical Trial
by promoting fibrosis at least in part through EMT, which itself in Overt Type 1 Diabetic Nephropathy), a phase-III clinical
has been correlated with the extent and severity of intersti- trial, failed to demonstrate any significant differences between
tial fibrosis and renal failure [77]. The postulated mechanisms placebo and aminoguanidine-treated groups, and only a trend
of senescence and ageing in organisms, ranging from yeast to towards lower serum creatinine and lower urinary protein was
mammals, include environmental and genetic factors, elevated observed with the treatment. The study was stopped because
oxidative stress, cumulative DNA damage, altered gene expres- of severe adverse effects such as abnormal liver function tests,
sion, telomere shortening and energy utilization. As AGEs are gastrointestinal disturbances, flu-like symptoms and vasculitis
implicated in physiological ageing, diabetes might be consid- or lupus-like phenomena [91].
ered the equivalent of accelerated ageing, particularly at the Another AGE inhibitor has also been studied—namely,
level of the vascular tree [7]. AGE blood levels and tissue accu- 2-isopropylidenehydrazono-4-oxo-thiazolidin-5-ylacetalinide
mulation are increased in organs (blood vessels, brain, eyes (OPB-9195), an ROS chelator that prevents the formation
and kidneys) in healthy elderly people [21,78–80], and AGEs of glucose derivatives such as methylglyoxal, glyoxal and 3-
also accumulate in various tissues with ageing. In Alzheimer’s deoxyglucosone, intermediaries that are indispensable for AGE
disease, AGEs have been found in neurofibrillary tangles and ␤- formation [93,94]. OPB-9195 is able to slow the progression
amyloid plaques [81,82]. AGEs also have direct cellular toxicity of glomerulosclerosis through a reduction in glycated collagen.
and are able to induce apoptosis through the caspase pathway However, clinical studies were stopped because of vitamin B6
[83,84]. As glycation of tau is followed by reduction of its sol- deficiency induced by OPB-9195 (pyridoxal chelator).
ubility and ability to be degraded, this may represent one of Of the aromatic ‘LR’ group, LR-90, LR-9 and LR-74 all
the first steps in the development of Alzheimer’s [85]. Also, as inhibit glycation of Amadori products and block glycoxidation.
many cells and tissues of the eye are profoundly influenced by Their in vivo effects are comparable to those of aminoguani-
glycation, AGEs are now receiving considerable attention as a dine and OPB-9195 [95]. Pyridoxamine, a vitamin B6 derivative,
possible pathogenic factor in vision disorders. AGEs accumulate inhibits the transformation of intermediary glycation products
in the macula in age-related macular degeneration, and RAGE [96]. Following injection of pyridoxamine in diabetic rats, a
activation in retinal pigment epithelial cells may contribute to decrease in the progression of DN has been observed [97]. Pyri-
the up-regulation of VEGF, thereby potentially inciting or prop- doxamine has also been used in phase-II clinical trials without
agating neovascular macular disease [86]. In addition, numerous major side-effects [98,99]. Furthermore, benfotiamine, a liposol-
factors, including AGEs, can contribute to renal ageing. Through uble vitamin B1 derivative, acts on glucose-derived products and
AGE accumulation, in situ glycation and RAGE activation, gly- reduces the glycation of albumin [100]. Benfotiamine can reduce
cation could promote the physiological and pathological effects AGE formation in endothelial cells and prevent the development
of renal ageing. It has recently been shown that AGEs in the of DN lesions [101]. In type 2 diabetic patients, oral benfoti-
diet correlate with serum AGE levels, oxidative stress, lifespan amine prevents endothelial dysfunction induced by a rich AGE
and renal dysfunction. Indeed, exogenous (dietary) AGEs are diet [102].
reported to accelerate ageing in mice, while the addition of a There are several possible ways to limit tissue AGE accu-
chemically defined AGE to low-AGE mouse chow can predis- mulation and reduce their deleterious effects. Alagebrium
pose to the development of cardiovascular and chronic kidney (ALT-711) is an AGE cross-link breaker (sugar/protein cross-
diseases [87]. link) that can reduce AGE accumulation and improve endothelial
dysfunction [103]. ALT-711 administration in diabetic mice pre-
5. Prevention and treatment vented the biological and histological changes of DN [104].
Several clinical trials have also demonstrated beneficial effects
Diet may play a key role in the prevention of AGE-linked of ALT-711 on blood pressure, arterial stiffness, left ventricular
pathologies. Food acts as a dietary source of variable amounts mass and diastolic filling [105–107].
of exogenous AGE, depending on its sugar and protein compo- In addition, a number of treatments currently in use as tar-
sition, and how it is cooked. The richer in proteins and sugars geted therapies for various disorders have recently been reported
a food is, and the more it is heated, the more AGEs it will con- to have ‘anti-AGE’ properties, including renin–angiotensin–
tain. In murine models, food restriction or a diet low in AGE aldosterone system blockers [angiotensin-converting enzyme
protects against the development of renal lesions [87,88]. Main- (ACE) inhibitors, angiotensin II receptor blockers (ARBs)],
taining optimal glycaemic balance provides a way of limiting statins, some oral antidiabetics and antioxidative molecules. In
endogenous AGE neoformation. vitro, ACE inhibitors and ARBs are able to block the secretion
Several targeted molecules have been tested as AGE of AGE-induced TGF-␤ and reduce the formation of ROS [108].
inhibitors. Aminoguanidine interacts with glucose-derived These agents can also increase blood concentrations of sRAGE,
products obtained from glycoxidation and the polyol pathway. which can bind AGEs, thereby reducing renal AGE deposits and
Its beneficial effect has, to a great extent, been demonstrated DN progression [109,110], and have beneficial effects on oxida-
in vitro and in vivo in animal models, with significant reduction tive stress, reducing the glycoxidation pathway [111]. Statins can
of AGE levels in blood and tissues [89,90]. In diabetic mice, reduce RAGE expression in diabetic patients, leading to effects
aminoguanidine not only prevents DN, but also diabetic on MCP-1 production [112]. In vitro, metformin and piogli-
M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10 7

tazone were able to reduce AGE formation, but these effects putative role in the development of chronic renal transplant dysfunction.
are yet to be confirmed in vivo [113–115]. Furthermore, some Am J Kidney Dis 2004;43:966–75.
antioxidants have demonstrated antiglycation effects [116]. [6] Berrou J, Tostivint I, Verrecchia F, Berthier C, Boulanger E, Mauviel A, et
al. Advanced glycation end-products regulate extracellular matrix protein
Other compounds that are currently only used for basic and protease expression by human glomerular mesangial cells. Int J Mol
research may be developed for therapeutic purposes. The most Med 2009;23:513–20.
promising candidates are sRAGE and anti-RAGE antibody. [7] Boulanger E, Puisieux F, Gaxatte C, Wautier JL. Aging: role and control
sRAGE acts as a decoy by binding AGEs and blocking RAGE of glycation. Rev Med Interne 2007;28:832–40.
activation and, when injected intraperitoneally into db/db mice, [8] Maillard L. Action des acides aminés sur les sucres ; formation des
mélanoïdes par voie méthodique. C R Acad Sci 1912:66–8.
prevents albuminuria, glomerulosclerosis and glomerular base- [9] Ohgami N, Nagai R, Ikemoto M, Arai H, Miyazaki A, Hakamata H, et al.
ment membrane (GBM) thickening [49]. Administration of CD36, serves as a receptor for advanced glycation end-products (AGE).
sRAGE also reduces inflammatory factors such as VCAM-1, TF J Diabetes Complications 2002;16:56–9.
and TGF-␤1, and stabilizes atherosclerotic lesions [117,118]. [10] Vlassara H, Li YM, Imani F, Wojciechowicz D, Yang Z, Liu FT, et al.
Anti-RAGE antibody specifically blocks RAGE activation. In Identification of galectin-3 as a high-affinity binding protein for advanced
glycation end-products (AGE): a new member of the AGE-receptor com-
vitro, RAGE antibodies reduce ECM remodelling [6]. Admin- plex. Mol Med 1995;1:634–46.
istration of anti-RAGE antibody over a 2-week period reduced [11] Schmidt AM, Vianna M, Gerlach M, Brett J, Ryan J, Kao J, et al. Isolation
serum creatinine, albuminuria, GBM thickness and mesangial and characterization of two binding proteins for advanced glycosylation
volume in db/db and type 1 diabetic mice [119,120]. end-products from bovine lung which are present on the endothelial cell
surface. J Biol Chem 1992;267:14987–97.
[12] Stern D, Yan SD, Yan SF, Schmidt AM. Receptor for advanced glycation
6. Conclusion end-products: a multiligand receptor magnifying cell stress in diverse
pathologic settings. Adv Drug Deliv Rev 2002;54:1615–25.
AGEs have numerous implications in ‘diabetic [13] Wautier JL, Guillausseau PJ. Advanced glycation end-products,
nephropathies’. However, although they play a major role their receptors and diabetic angiopathy. Diabetes Metab 2001;27:
in the development of DN itself, their presence has also 535–42.
[14] Sugaya K, Fukagawa T, Matsumoto K, Mita K, Takahashi E, Ando A, et
been demonstrated in other renal diseases and in ageing. The al. Three genes in the human MHC class III region near the junction with
battle against AGEs constitutes an integral part of the current the class II: gene for receptor of advanced glycosylation end-products.
recommendations for renoprotective and ‘future’ anti-ageing PBX2 homeobox gene and a notch homolog, human counterpart of mouse
strategies. Nevertheless, while awaiting the development of a mammary tumor gene int-3. Genomics 1994;23:408–19.
patient-specific, targeted therapeutic approach, there is still a [15] Kalea AZ, Schmidt AM, Hudson BI. RAGE: a novel biological and
genetic marker for vascular disease. Clin Sci 2009;116:621–37.
need for strict glycaemic control, a suitably adapted diet, and [16] Pettersson-Fernholm K, Forsblom C, Hudson BI, Perola M, Grant PJ,
vascular protection through the use of ACE inhibitors or ARBs, Groop PH. The functional -374 T/A RAGE gene polymorphism is asso-
statins and certain oral antidiabetic agents. ciated with proteinuria and cardiovascular disease in type 1 diabetic
patients. Diabetes 2003;52:891–4.
7. Conflicts of interest [17] Prevost G, Fajardy I, Besmond C, Balkau B, Tichet J, Fontaine P, et
al. Polymorphisms of the receptor of advanced glycation end-products
(RAGE) and the development of nephropathy in type 1 diabetic patients.
No potential conflicts of interest relevant to this article have Diabetes Metab 2005;31:35–9.
been reported. [18] Hudson BI, Carter AM, Harja E, Kalea AZ, Arriero M, Yang H, et al. Iden-
tification, classification, and expression of RAGE gene splice variants.
Acknowledgments FASEB J 2008;22:1572–80.
[19] Raucci A, Cugusi S, Antonelli A, Barabino SM, Monti L, Bierhaus A, et
al. A soluble form of the receptor for advanced glycation end-products
Pr Éric Boulanger has held a post-doctoral position in the (RAGE) is produced by proteolytic cleavage of the membrane-bound
laboratory of Dr Ann Marie Schmidt at Columbia University in form by the sheddase a disintegrin and metalloprotease 10 (ADAM10).
NY, USA, and would like to thank ALFEDIAM (Association FASEB J 2008;22:3716–27.
de langue française pour l’étude du diabète et des maladies [20] Brett J, Schmidt AM, Yan SD, Zou YS, Weidman E, Pinsky D, et al.
Survey of the distribution of a newly characterized receptor for advanced
métaboliques) for kindly providing him with a grant. glycation end-products in tissues. Am J Pathol 1993;143:1699–712.
[21] Soulis T, Thallas V, Youssef S, Gilbert RE, McWilliam BG, Murray-
References McIntosh RP, et al. Advanced glycation end-products and their receptors
co-localise in rat organs susceptible to diabetic microvascular injury.
[1] Dikow R, Ritz E. In: Davidson AM, Stewart Cameron J, Grünfeld JP, Diabetologia 1997;40:619–28.
Ponticelli C, Ritz E, Winearls CG, et al., editors. The patient with diabetes [22] Boulanger E, Wautier MP, Wautier JL, Boval B, Panis Y, Wernert N, et
mellitus. International textbook of clinical nephrology; 2006. p. 661–73. al. AGEs bind to mesothelial cells via RAGE and stimulate VCAM-1
[2] Miyata T. New aspects in the pathogenesis of dialysis-related amyloido- expression. Kidney Int 2002;61:148–56.
sis: pathophysiology of advanced glycation end-products in renal failure. [23] Tanji N, Markowitz GS, Fu C, Kislinger T, Taguchi A, Pischetsrieder M,
Nippon Jinzo Gakkai Shi 1996;38:191–7. et al. Expression of advanced glycation end-products and their cellular
[3] Simonson MS. Phenotypic transitions and fibrosis in diabetic nephropa- receptor RAGE in diabetic nephropathy and nondiabetic renal disease. J
thy. Kidney Int 2007;71:846–54. Am Soc Nephrol 2000;11:1656–66.
[4] Bohlender JM, Franke S, Stein G, Wolf G. Advanced glycation end- [24] Suzuki D, Toyoda M, Yamamoto N, Miyauchi M, Katoh M, Kimura M,
products and the kidney. Am J Physiol Renal Physiol 2005;289:645–59. et al. Relationship between the expression of advanced glycation end-
[5] Hartog JW, Smit AJ, van Son WJ, Navis G, Gans RO, Wolffenbuttel BH, products (AGE) and the receptor for AGE (RAGE) mRNA in diabetic
et al. Advanced glycation end-products in kidney transplant patients: a nephropathy. Intern Med 2006;45:435–41.
8 M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10

[25] Wautier MP, Chappey O, Corda S, Stern DM, Schmidt AM, Wautier JL. p21waf to control cell-cycle progression of mesangial cells: a possible
Activation of NADPH oxidase by AGE links oxidant stress to altered gene role in the development and progression of diabetic nephropathy. FASEB
expression via RAGE. Am J Physiol Endocrinol Metab 2001;280:685–94. J 2004;18:1249–51.
[26] Schmidt AM, Yan SD, Yan SF, Stern DM. The multiligand receptor RAGE [44] Yamagishi S, Inagaki Y, Okamoto T, Amano S, Koga K, Takeuchi
as a progression factor amplifying immune and inflammatory responses. M, et al. Advanced glycation end-product-induced apoptosis and
J Clin Invest 2001;108:949–55. overexpression of vascular endothelial growth factor and monocyte
[27] Wautier JL, Schmidt AM. Protein glycation: a firm link to endothelial cell chemoattractant protein-1 in human-cultured mesangial cells. J Biol
dysfunction. Circ Res 2004;95:233–8. Chem 2002;277:20309–15.
[28] Boulanger E, Grossin N, Wautier MP, Taamma R, Wautier JL, Mesothe- [45] Ha H, Lee HB. Reactive oxygen species as glucose signaling
lial. RAGE activation by AGEs enhances VEGF release and potentiates molecules in mesangial cells cultured under high glucose. Kidney Int
capillary tube formation. Kidney Int 2007;71:126–33. 2000;77(Suppl.):S19–25.
[29] Lo CS, Relf IR, Myers KA, Wahlqvist ML. Doppler ultrasound recogni- [46] Li YM, Mitsuhashi T, Wojciechowicz D, Shimizu N, Li J, Stitt A,
tion of preclinical changes in arterial wall in diabetic subjects: compliance et al. Molecular identity and cellular distribution of advanced glyca-
and pulse-wave damping. Diabetes Care 1986;9:27–31. tion end-product receptors: relationship of p60 to OST-48 and p90
[30] Stitt AW, Bucala R, Vlassara H. Atherogenesis and advanced gly- to 80K-H membrane proteins. Proc Natl Acad Sci U S A 1996;93:
cation: promotion, progression, and prevention. Ann N Y Acad Sci 11047–52.
1997;811:115–27. [47] He CJ, Zheng F, Stitt A, Striker L, Hattori M, Vlassara H. Differential
[31] Yamada K, Miyahara Y, Hamaguchi K, Nakayama M, Nakano H, expression of renal AGE-receptor genes in NOD mice: possible role in
Nozaki O, et al. Immunohistochemical study of human advanced gly- nonobese diabetic renal disease. Kidney Int 2000;58:1931–40.
cosylation end-products (AGE) in chronic renal failure. Clin Nephrol [48] Wendt T, Tanji N, Guo J, Hudson BI, Bierhaus A, Ramasamy R,
1994;42:354–61. et al. Glucose, glycation, and RAGE: implications for amplification
[32] Gugliucci A, Bendayan M. Reaction of advanced glycation end-products of cellular dysfunction in diabetic nephropathy. J Am Soc Nephrol
with renal tissue from normal and streptozotocin-induced diabetic rats: 2003;14:1383–95.
an ultrastructural study using colloidal gold cytochemistry. J Histochem [49] Wendt TM, Tanji N, Guo J, Kislinger TR, Qu W, Lu Y, et al. RAGE
Cytochem 1995;43:591–600. drives the development of glomerulosclerosis and implicates podocyte
[33] Horie K, Miyata T, Maeda K, Miyata S, Sugiyama S, Sakai H, et al. activation in the pathogenesis of diabetic nephropathy. Am J Pathol
Immunohistochemical colocalization of glycoxidation products and lipid 2003;162:1123–37.
peroxidation products in diabetic renal glomerular lesions. Implication [50] Chuang PY, Yu Q, Fang W, Uribarri J, He JC. Advanced glycation
for glycoxidative stress in the pathogenesis of diabetic nephropathy. J end-products induce podocyte apoptosis by activation of the FOXO4
Clin Invest 1997;100:2995–3004. transcription factor. Kidney Int 2007;72:965–76.
[34] Wautier MP, Boulanger E, Guillausseau PJ, Massin P, Wautier JL. AGEs, [51] Doublier S, Salvidio G, Lupia E, Ruotsalainen V, Verzola D, Deferrari G,
macrophage colony stimulating factor and vascular adhesion molecule et al. Nephrin expression is reduced in human diabetic nephropathy: evi-
blood levels are increased in patients with diabetic microangiopathy. dence for a distinct role for glycated albumin and angiotensin II. Diabetes
Thromb Haemost 2004;91:879–85. 2003;52:1023–30.
[35] Basta G, Lazzerini G, Massaro M, Simoncini T, Tanganelli P, Fu C, [52] Lachapelle M, Bendayan M. Contractile proteins in podocytes: immuno-
et al. Advanced glycation end-products activate endothelium through cytochemical localization of actin and alpha-actinin in normal and
signal-transduction receptor RAGE: a mechanism for amplification of nephrotic rat kidneys. Virchows Arch B Cell Pathol Incl Mol Pathol
inflammatory responses. Circulation 2002;105:816–22. 1991;60:105–11.
[36] Schmidt AM, Hori O, Chen JX, Li JF, Crandall J, Zhang J, et al. Advanced [53] Ha TS. High glucose and advanced glycosylated end-products affect the
glycation end-products interacting with their endothelial receptor induce expression of alpha-actinin-4 in glomerular epithelial cells. Nephrology
expression of vascular cell adhesion molecule-1 (VCAM-1) in cultured 2006;11:435–41.
human endothelial cells and in mice. A potential mechanism for the [54] Kimura M, Toyoda M, Kato M, Kobayashi K, Abe M, Kobayashi T, et
accelerated vasculopathy of diabetes. J Clin Invest 1995;96:1395–403. al. Expression of alpha-actinin-4 in human diabetic nephropathy. Intern
[37] Giardino I, Edelstein D, Brownlee M. Nonenzymatic glycosylation in Med 2008;47:1099–106.
vitro and in bovine endothelial cells alters basic fibroblast growth factor [55] Li JH, Wang W, Huang XR, Oldfield M, Schmidt AM, Cooper ME, et al.
activity. A model for intracellular glycosylation in diabetes. J Clin Invest Advanced glycation end-products induce tubular epithelial-myofibroblast
1994;94:110–7. transition through the RAGE-ERK1/2 MAP kinase signaling pathway.
[38] Xu B, Ji Y, Yao K, Cao YX, Ferro A. Inhibition of human endothelial Am J Pathol 2004;164:1389–97.
cell nitric oxide synthesis by advanced glycation end-products but not [56] Oldfield MD, Bach LA, Forbes JM, Nikolic-Paterson D, McRobert
glucose: relevance to diabetes. Clin Sci 2005;109:439–46. A, Thallas V, et al. Advanced glycation end-products cause epithelial-
[39] Bucala R, Tracey KJ, Cerami A. Advanced glycosylation products quench myofibroblast transdifferentiation via the receptor for advanced glycation
nitric oxide and mediate defective endothelium-dependent vasodilatation end-products (RAGE). J Clin Invest 2001;108:1853–63.
in experimental diabetes. J Clin Invest 1991;87:432–8. [57] Reiser K, McCormick RJ, Rucker RB. Enzymatic and nonen-
[40] Esposito C, Gerlach H, Brett J, Stern D, Vlassara H. Endothelial zymatic cross-linking of collagen and elastin. FASEB J 1992;6:
receptor-mediated binding of glucose-modified albumin is associated 2439–49.
with increased monolayer permeability and modulation of cell surface [58] McLennan SV, Fisher EJ, Yue DK, Turtle JR. High glucose concentration
coagulant properties. J Exp Med 1989;170:1387–407. causes a decrease in mesangium degradation. A factor in the pathogenesis
[41] Wautier JL, Zoukourian C, Chappey O, Wautier MP, Guillausseau of diabetic nephropathy. Diabetes 1994;43:1041–5.
PJ, Cao R, et al. Receptor-mediated endothelial cell dysfunction in [59] Mott JD, Khalifah RG, Nagase H, Shield 3rd CF, Hudson JK, Hudson BG.
diabetic vasculopathy. Soluble receptor for advanced glycation end- Nonenzymatic glycation of type IV collagen and matrix metalloproteinase
products blocks hyperpermeability in diabetic rats. J Clin Invest 1996;97: susceptibility. Kidney Int 1997;52:1302–12.
238–43. [60] McLennan SV, Martell SK, Yue DK. Effects of mesangium glycation on
[42] Bulckaen H, Prevost G, Boulanger E, Robitaille G, Roquet V, Gaxatte C, matrix metalloproteinase activities: possible role in diabetic nephropathy.
et al. Low-dose aspirin prevents age-related endothelial dysfunction in a Diabetes 2002;51:2612–8.
mouse model of physiological aging. Am J Physiol Heart Circ Physiol [61] McLennan SV, Kelly DJ, Schache M, Waltham M, Dy V, Langham RG,
2008;294:1562–70. et al. Advanced glycation end-products decrease mesangial cell MMP-
[43] Brizzi MF, Dentelli P, Rosso A, Calvi C, Gambino R, Cassader M, et al. 7: a role in matrix accumulation in diabetic nephropathy? Kidney Int
RAGE- and TGF-beta receptor-mediated signals converge on STAT5 and 2007;72:481–8.
M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10 9

[62] Krishnamurti U, Rondeau E, Sraer JD, Michael AF, Tsilibary EC. ney function in older community-dwelling women. Am J Kidney Dis
Alterations in human glomerular epithelial cells interacting with nonen- 2009;53:51–8.
zymatically glycosylated matrix. J Biol Chem 1997;272:27966–70. [81] Reddy VP, Obrenovich ME, Atwood CS, Perry G, Smith MA. Involve-
[63] Vlassara H, Striker LJ, Teichberg S, Fuh H, Li YM, Steffes M. Advanced ment of Maillard reactions in Alzheimer disease. Neurotox Res
glycation end-products induce glomerular sclerosis and albuminuria in 2002;4:191–209.
normal rats. Proc Natl Acad Sci U S A 1994;91:11704–8. [82] Wong A, Luth HJ, Deuther-Conrad W, Dukic-Stefanovic S, Gasic-
[64] Muller-Krebs S, Kihm LP, Zeier B, Gross ML, Deppisch R, Wieslander Milenkovic J, Arendt T, et al. Advanced glycation end-products
A, et al. Renal toxicity mediated by glucose degradation products in a rat co-localize with inducible nitric oxide synthase in Alzheimer’s disease.
model of advanced renal failure. Eur J Clin Invest 2008;38:296–305. Brain Res 2001;920:32–40.
[65] Miyata T, Ueda Y, Yamada Y, Izuhara Y, Wada T, Jadoul M, et al. Accumu- [83] Artwohl M, Graier WF, Roden M, Bischof M, Freudenthaler A, Waldhausl
lation of carbonyls accelerates the formation of pentosidine, an advanced W, et al. Diabetic LDL triggers apoptosis in vascular endothelial cells.
glycation end-product: carbonyl stress in uremia. J Am Soc Nephrol Diabetes 2003;52:1240–7.
1998;9:2349–56. [84] Luth HJ, Ogunlade V, Kuhla B, Kientsch-Engel R, Stahl P, Webster J,
[66] Foerster A, Henle T. Glycation in food and metabolic transit of dietary et al. Age- and stage-dependent accumulation of advanced glycation
AGEs (advanced glycation end-products): studies on the urinary excretion end-products in intracellular deposits in normal and Alzheimer’s disease
of pyrraline. Biochem Soc Trans 2003;31:1383–5. brains. Cereb Cortex 2005;15:211–20.
[67] Uribarri J, Peppa M, Cai W, Goldberg T, Lu M, He C, et al. Restriction of [85] Munch G, Luth HJ, Wong A, Arendt T, Hirsch E, Ravid R, et al. Crosslink-
dietary glycotoxins reduces excessive advanced glycation end-products ing of alpha-synuclein by advanced glycation end-products: an early
in renal failure patients. J Am Soc Nephrol 2003;14:728–31. pathophysiological step in Lewy body formation? J Chem Neuroanat
[68] Uribarri J, Peppa M, Cai W, Goldberg T, Lu M, Baliga S, et al. Dietary 2000;20:253–7.
glycotoxins correlate with circulating advanced glycation end-product [86] Ma W, Lee SE, Guo J, Qu W, Hudson BI, Schmidt AM, et al. RAGE ligand
levels in renal failure patients. Am J Kidney Dis 2003;42:532–8. upregulation of VEGF secretion in ARPE-19 cells. Invest Ophthalmol Vis
[69] Gugliucci A, Bendayan M. Renal fate of circulating advanced gly- Sci 2007;48:1355–61.
cated end-products (AGE): evidence for reabsorption and catabolism [87] Cai W, He JC, Zhu L, Chen X, Zheng F, Striker GE, et al. Oral glycotoxins
of AGE-peptides by renal proximal tubular cells. Diabetologia 1996;39: determine the effects of calorie restriction on oxidant stress, age-related
149–60. diseases, and lifespan. Am J Pathol 2008;173:327–36.
[70] Kostolanska J, Jakus V, Barak L. Monitoring of early and advanced [88] Teillet L, Verbeke P, Gouraud S, Bakala H, Borot-Laloi C, Heudes D, et al.
glycation in relation to the occurrence of microvascular complications Food restriction prevents advanced glycation end-product accumulation
in children and adolescents with type 1 diabetes mellitus. Physiol Res and retards kidney aging in lean rats. J Am Soc Nephrol 2000;11:1488–97.
2009;58:553–61. [89] Soulis T, Cooper ME, Vranes D, Bucala R, Jerums G. Effects of
[71] Guo J, Ananthakrishnan R, Qu W, Lu Y, Reiniger N, Zeng S, et al. RAGE aminoguanidine in preventing experimental diabetic nephropathy are
mediates podocyte injury in adriamycin-induced glomerulosclerosis. J related to the duration of treatment. Kidney Int 1996;50:627–34.
Am Soc Nephrol 2008;19:961–72. [90] Kelly DJ, Gilbert RE, Cox AJ, Soulis T, Jerums G, Cooper ME.
[72] Kilhovd BK, Juutilainen A, Lehto S, Ronnemaa T, Torjesen PA, Birke- Aminoguanidine ameliorates overexpression of prosclerotic growth fac-
land KI, et al. High serum levels of advanced glycation end-products tors and collagen deposition in experimental diabetic nephropathy. J Am
predict increased coronary heart disease mortality in nondiabetic women Soc Nephrol 2001;12:2098–107.
but not in nondiabetic men: a population-based 18-year follow-up study. [91] Peyroux J, Sternberg M. Advanced glycation end-products (AGEs): phar-
Arterioscler Thromb Vasc Biol 2005;25:815–20. macological inhibition in diabetes. Pathol Biol 2006;54:405–19.
[73] Suzuki D, Miyata T, Saotome N, Horie K, Inagi R, Yasuda Y, et al. [92] Huijberts MS, Wolffenbuttel BH, Boudier HA, Crijns FR, Kruseman
Immunohistochemical evidence for an increased oxidative stress and car- AC, Poitevin P, et al. Aminoguanidine treatment increases elasticity and
bonyl modification of proteins in diabetic glomerular lesions. J Am Soc decreases fluid filtration of large arteries from diabetic rats. J Clin Invest
Nephrol 1999;10:822–32. 1993;92:1407–11.
[74] de Leeuw K, Graaff R, de Vries R, Dullaart RP, Smit AJ, Kallenberg CG, [93] Nakamura S, Makita Z, Ishikawa S, Yasumura K, Fujii W, Yanagisawa
et al. Accumulation of advanced glycation end-products in patients with K, et al. Progression of nephropathy in spontaneous diabetic rats is pre-
systemic lupus erythematosus. Rheumatology 2007;46:1551–6. vented by OPB-9195, a novel inhibitor of advanced glycation. Diabetes
[75] Qing X, Pitashny M, Thomas DB, Barrat FJ, Hogarth MP, Putterman C. 1997;46:895–9.
Pathogenic anti-DNA antibodies modulate gene expression in mesangial [94] Tsuchida K, Makita Z, Yamagishi S, Atsumi T, Miyoshi H, Obara S, et al.
cells: involvement of HMGB1 in anti-DNA antibody-induced renal injury. Suppression of transforming growth factor beta and vascular endothelial
Immunol Lett 2008;121:61–73. growth factor in diabetic nephropathy in rats by a novel advanced glyca-
[76] Linden E, Cai W, He JC, Xue C, Li Z, Winston J, et al. Endothe- tion end-product inhibitor, OPB-9195. Diabetologia 1999;42:579–88.
lial dysfunction in patients with chronic kidney disease results from [95] Wolffenbuttel BH, Boulanger CM, Crijns FR, Huijberts MS, Poitevin P,
advanced glycation end-products (AGE)-mediated inhibition of endothe- Swennen GN, et al. Breakers of advanced glycation end-products restore
lial nitric oxide synthase through RAGE activation. Clin J Am Soc large artery properties in experimental diabetes. Proc Natl Acad Sci U S
Nephrol 2008;3:691–8. A 1998;95:4630–4.
[77] Lan HY. Tubular epithelial-myofibroblast transdifferentiation mech- [96] Booth AA, Khalifah RG, Todd P, Hudson BG. In vitro kinetic stud-
anisms in proximal tubule cells. Curr Opin Nephrol Hypertens ies of formation of antigenic advanced glycation end-products (AGEs).
2003;12:25–9. Novel inhibition of post-Amadori glycation pathways. J Biol Chem
[78] Vitek MP, Bhattacharya K, Glendening JM, Stopa E, Vlassara H, 1997;272:5430–7.
Bucala R, et al. Advanced glycation end-products contribute to amy- [97] Degenhardt TP, Alderson NL, Arrington DD, Beattie RJ, Basgen JM,
loidosis in Alzheimer disease. Proc Natl Acad Sci U S A 1994;91: Steffes MW, et al. Pyridoxamine inhibits early renal disease and dyslipi-
4766–70. demia in the streptozotocin-diabetic rat. Kidney Int 2002;61:939–50.
[79] Glenn JV, Beattie JR, Barrett L, Frizzell N, Thorpe SR, Boulton ME, [98] Williams ME. New therapies for advanced glycation end-product nephro-
et al. Confocal Raman microscopy can quantify advanced glycation toxicity: current challenges. Am J Kidney Dis 2003;41:S42–7.
end-product (AGE) modifications in Bruch’s membrane leading to accu- [99] Voziyan PA, Hudson BG. Pyridoxamine as a multifunctional pharmaceu-
rate, nondestructive prediction of ocular aging. FASEB J 2007;21: tical: targeting pathogenic glycation and oxidative damage. Cell Mol Life
3542–52. Sci 2005;62:1671–81.
[80] Semba RD, Ferrucci L, Fink JC, Sun K, Beck J, Dalal M, et al. Advanced [100] Berrone E, Beltramo E, Solimine C, Ape AU, Porta M. Regulation
glycation end-products and their circulating receptors and level of kid- of intracellular glucose and polyol pathway by thiamine and benfoti-
10 M. Daroux et al. / Diabetes & Metabolism 36 (2010) 1–10

amine in vascular cells cultured in high glucose. J Biol Chem 2006;281: and AGE inhibition in the diabetic spontaneously hypertensive rat. Dia-
9307–13. betologia 2004;47:89–97.
[101] Hammes HP, Du X, Edelstein D, Taguchi T, Matsumura T, Ju Q, et al. [111] Miyata T, van Ypersele de Strihou C, Ueda Y, Ichimori K, Inagi R, Onogi
Benfotiamine blocks three major pathways of hyperglycemic damage and H, et al. Angiotensin II receptor antagonists and angiotensin-converting
prevents experimental diabetic retinopathy. Nat Med 2003;9:294–9. enzyme inhibitors lower in vitro the formation of advanced glycation
[102] Stirban A, Negrean M, Stratmann B, Gawlowski T, Horstmann T, Got- end-products: biochemical mechanisms. J Am Soc Nephrol 2002;13:
ting C, et al. Benfotiamine prevents macro- and microvascular endothelial 2478–87.
dysfunction and oxidative stress following a meal rich in advanced gly- [112] Cuccurullo C, Iezzi A, Fazia ML, De Cesare D, Di Francesco A, Muraro
cation end-products in individuals with type 2 diabetes. Diabetes Care R, et al. Suppression of RAGE as a basis of simvastatin-dependent
2006;29:2064–71. plaque stabilization in type 2 diabetes. Arterioscler Thromb Vasc Biol
[103] Coughlan MT, Forbes JM, Cooper ME. Role of the AGE crosslink 2006;26:2716–23.
breaker, alagebrium, as a renoprotective agent in diabetes. Kidney Int [113] Ruggiero-Lopez D, Lecomte M, Moinet G, Patereau G, Lagarde M,
2007;106(Suppl.):S54–60. Wiernsperger N. Reaction of metformin with dicarbonyl compounds.
[104] Peppa M, Brem H, Cai W, Zhang JG, Basgen J, Li Z, et al. Prevention and Possible implication in the inhibition of advanced glycation end-product
reversal of diabetic nephropathy in db/db mice treated with alagebrium formation. Biochem Pharmacol 1999;58:1765–73.
(ALT-711). Am J Nephrol 2006;26:430–6. [114] Scheen AJ, Paquot N, Lefebvre PJ. United Kingdom Prospective
[105] Little WC, Zile MR, Kitzman DW, Hundley WG, O’Brien TX, Degroof Diabetes Study (UKPDS): 10 years later. Rev Med Liege 2008;63:
RC. The effect of alagebrium chloride (ALT-711), a novel glucose cross- 624–9.
link breaker, in the treatment of elderly patients with diastolic heart failure. [115] Rahbar S, Natarajan R, Yerneni K, Scott S, Gonzales N, Nadler JL. Evi-
J Card Fail 2005;11:191–5. dence that pioglitazone, metformin and pentoxifylline are inhibitors of
[106] Vasan S, Foiles P, Founds H. Therapeutic potential of breakers of glycation. Clin Chim Acta 2000;301:65–77.
advanced glycation end-product-protein crosslinks. Arch Biochem Bio- [116] Bonnefont-Rousselot D. Antioxidant and anti-AGE therapeutics: evalua-
phys 2003;419:89–96. tion and perspectives. J Soc Biol 2001;195:391–8.
[107] Kass DA, Shapiro EP, Kawaguchi M, Capriotti AR, Scuteri A, [117] Kislinger T, Tanji N, Wendt T, Qu W, Lu Y, Ferran Jr LJ, et al. Receptor for
deGroof RC, et al. Improved arterial compliance by a novel advanced glycation end-products mediates inflammation and enhanced
advanced glycation end-product crosslink breaker. Circulation 2001;104: expression of tissue factor in vasculature of diabetic apolipoprotein E-null
1464–70. mice. Arterioscler Thromb Vasc Biol 2001;21:905–10.
[108] Fukami K, Ueda S, Yamagishi S, Kato S, Inagaki Y, Takeuchi M, et al. [118] Bucciarelli LG, Wendt T, Qu W, Lu Y, Lalla E, Rong LL, et al. RAGE
AGEs activate mesangial TGF-beta-Smad signaling via an angiotensin II blockade stabilizes established atherosclerosis in diabetic apolipoprotein
type I receptor interaction. Kidney Int 2004;66:2137–47. E-null mice. Circulation 2002;106:2827–35.
[109] Forbes JM, Thorpe SR, Thallas-Bonke V, Pete J, Thomas MC, Deemer [119] Flyvbjerg A, Denner L, Schrijvers BF, Tilton RG, Mogensen TH, Paludan
ER, et al. Modulation of soluble receptor for advanced glycation end- SR, et al. Long-term renal effects of a neutralizing RAGE antibody in
products by angiotensin-converting enzyme-1 inhibition in diabetic obese type 2 diabetic mice. Diabetes 2004;53:166–72.
nephropathy. J Am Soc Nephrol 2005;16:2363–72. [120] Jensen LJ, Denner L, Schrijvers BF, Tilton RG, Rasch R, Flyvb-
[110] Davis BJ, Forbes JM, Thomas MC, Jerums G, Burns WC, Kawachi H, jerg A. Renal effects of a neutralising RAGE-antibody in long-term
et al. Superior renoprotective effects of combination therapy with ACE streptozotocin-diabetic mice. J Endocrinol 2006;188:493–501.

You might also like