Sie sind auf Seite 1von 6

Received: Dec 13, 2011

Accepted: May. 15, 2012


Published online: Jun. 30, 2012

Review Article

Advanced Glycation End Products and Their Receptors as Risk Factors for Aging
Ryoji Nagai 1), Masao Jinno 2), Masamitsu Ichihashi 3), Hidenori Koyama 4), Yasuhiko Yamamoto 5),
Yoshikazu Yonei 3)
1) Laboratory of Food and Regulation Biology Department of Bioscience, School of Agriculture, Tokai University
2) Womens Clinic Jinno
3) Anti-Aging Medical Research Center, Graduate School of Life and Medical Sciences, Doshisha University
4) Department of Endocrinology and Metabolism, Hyougo Medical College
5) Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Science

Abstract
Glycation is the reaction between amino residues of proteins and carbonyl of reducing sugars. French Louis Camille Maillard
discovers this reaction from the browning reaction by amino acid and sugar, and it is widely known for the food chemistry as the
Maillard reaction. The hemoglobin A1c (HbA1c) measured all over the world as a marker of glycemic control is equivalent to the
Amadori rearrangement products, which is the early stage product of the Maillard reaction. Therefore, glycation progresses even in
the healthy subject since carbohydrates are used as an energy source. Then, the Amadori products are changed to Advanced Glycation
End-products (AGEs), which shows the autofluorescence, browning, and cross-linking by oxidation, dehydration, and a condensation
reaction. Furthermore, AGE-proteins are recognized by AGE receptor such as RAGE (receptor for AGE). This review describes the
proposed Pathways for the formation of AGEs during the Maillard reaction and role of the AGEs in the pathogenesis of age-related
diseases.

KEY WORDS: AGEs, glycation, RAGE

Introduction

Maillard reaction (glycation reaction)

Despite the 100 or so years that have lapsed since French


scientist Louis-Camille Maillard first reported the Maillard
reaction between amino acids and glucose in 1912, in vivo
research has not progressed significantly. In recent years,
advances have been made in proteomics research, which is the
comprehensive analysis of expressed protein, thanks in large
part to the human genome project. However, this research
subsequently led to the identification of diseases which cannot
be cured simply by studying gene or protein expression. The
Maillard reaction has also recently attracted new attention for
its role as a posttranslational modification, promoted by an
abnormality in the metabolism of sugars and lipids. Advanced
glycation end products (AGEs) change the physicochemical
properties of proteins such as net negative chargedegeneration
and polymerization.
Recent studies have reported that formation of AGEs is
enhanced by not only abnormalities in the metabolism of sugar
but also by elevated oxidative stress. Further, the interaction
between AGEs and receptor for AGEs (RAGE) also cause
of inflammation and oxidative stress. Here, we describe the
biological significance of AGEs and RAGE as risk factors for
aging.

Taking its name from its discoverer, the Maillard reaction


is also known as glycation, as it progresses from the reaction
in which reducing sugar combines with protein. The reaction
can be divided into two parts: the first half proceeds until
Amadori rearrangement, and the second half involves AGE
formation through reactions such as oxidation, dehydration, and
condensation (Fig. 1). Food chemists have conducted substantial
research on this reaction since its initial discovery in relation to
food processing, browning by storage, and change in nutritive
value.
Hemoglobin A1c (HbA1c) was first identified in vivo in the
1970s, and subsequent research has revealed this molecule to
be the Amadori rearrangement product of glucose combining
with the N-terminal amino residue of valine on the hemoglobin
chain. While HbA1c is used globally as a marker for diabetic
glycemic control, clinical application of AGEs is limited by the
large number of AGE structures and the fact that measuring
methods differ by structure. However, as carbohydrates are
nutrients indispensable to energy production, an organism uses
these compounds throughout its life, and Maillard reactions
therefore progress as long as it is alive. Positively charged lysine
and arginine residues on proteins are modified by AGEs, and
increased net negative charge of proteins and forming crosslinkage structures are also known to influence the structure of
proteins, including the activity of enzymes (Fig. 2).

Anti-Aging Medicine 9 (4) : 108-113, 2012


(c) Japanese Society of Anti-Aging Medicine

108

Ryoji Nagai
Laboratory of Food and Regulation Biology Department of Bioscience, School of Agriculture, Tokai University
Kawayou, Minamiaso, Aso-gun, Kumamoto 869-1404, Japan
Tel & Fax: +81-967-67-3918 / E-mail: nagai-883@umin.ac.jp

Glycation as a Risk Factor for Aging

Fig. 1. Maillard reaction.

Fig. 2. Generation of intermediate aldehydes and protein degeneration involved in AGE formation.

revealing that AGE generation in living organisms involves not


only glucose but also various carbonyl compounds formed by
metabolism and inflammation reactions (Fig. 3), and that several
formation pathways exist for short-term rapid production of
AGEs 1,2). In recent years, a substantial amount of focus has been
directed towards the study of AGEs in the field of aging research,
as AGE formation is promoted in patients with lifestyle-related
diseases accelerated by aging, such as metabolic disorders
involving carbohydrates . Detailed measurements of certain
antibodies and instrumental analyses of several AGEs have also
accelerated the progress of research in this field.

In the past, measurement of f luorescence intensity has


primarily involved estimating the AGE content, which is a
relatively simple process. However, a number of concerns have
been raised regarding estimation of AGEs in vivo simply by
measuring fluorescence intensity, given the great number of
AGEs which do not fluoresce and the fact that some non-AGE
compounds show similar f luorescence properties to AGEs,
potentially confounding results.
In light of these drawbacks, assessment of AGE structures
based on anti-AGE antibodies has been widely employed, given
the inherent advantage with its multi-sample measurement
capabilities. In vivo AGE research has rapidly progressed as well,

109

Glycation as a Risk Factor for Aging

Fig. 3. Pathway of intermediate aldehyde generation.

pulmonary fibrosis caused by bleomycin exposure , osteoporosis,


sepsis, Alzheimers disease and colitis. Endogenous secretory
RAGE (esRAGE) is a secretory form of RAGE made by
alternative splicing of the RAGE gene 16), and soluble RAGE
(sRAGE) is formed by shedding full-length RAGE via activation
of enzymes such as MMP9 or ADAM10. Both esRAGE and
sRAGE have ligand-binding sites and work as decoy receptors by
capturing RAGE ligands 17).

Receptors for AGEs (RAGE)


Currently identified cell surface receptors which recognize
AGEs include RAGE; macrophage scavenger receptor class A
(SR-A); 3) SR-B (SR-B1 and CD36); 4-5) lectin-like oxidized low
density lipoprotein receptor-1 (LOX-1); 6) galectin-3 complex;
7) fasciclin , epidermal growth factor (EGF)-like, laminin-type
EGF-like, and link domain-containing scavenger receptors-1 and
-2 (FEEL-1 and -2); megalin; 8) and toll-like receptor 4 (TLR4)
(Fig. 4). RAGE and TLR4, both pattern-recognition receptors
(PRRs), are believed to be responsible for intracellular signal
transduction. RAGE is a type 1 transmembrane protein belonging
to the immunoglobulin superfamily and is known to be a multiligand receptor recognizing not only AGEs but also advanced
oxidation protein products (AOPPs) resulting from oxidative
stress 9), amyloid beta related to Alzheimers disease 10), highmobility group box-1 (HMGB-1) linked to cancer metastasis and
inflammation 11), inflammatory mediator S100 protein secreted
from immune cells 12), Mac1/CD11b on the cell surface of white
blood cells, lipopolysaccharide (LPS) of bacterial membrane
components 13), complement C3a, and phosphatidylserine on
apoptotic cells 14).
A typical intracellular signaling pathway of RAGE involves
formation of intracellular oxidation stress and activation of
transcription factor NFB. Studies in diabetic transgenic mice
overexpressing RAGE protein in vascular cells have shown that
the onset of diabetic nephropathy is accelerated in this model.
In contrast, studies in RAGE knockout mice with diabetes
have shown that nephropathy was ameliorated in these animals
15). Taken together, these previous findings demonstrate that
RAGE is functionally involved in the development of vascular
complications in diabetes.
In addition, RAGE-dependent inflammatory disorders were
demonstrated by animal models such as atherosclerosis, the

Current progress in AGE research


A number of studies have reported the marked accumulation
of the AGE structure N -(carboxymethyl)lysine (CML) in the
elastic fiber of solar elastosis 18), with related studies reporting a
clear relationship between CML formation and reactive oxygen
species (ROS) such as hydroxyl radicals 19), peroxynitrite 20),
and hypochlorous acid 21). In solar elastosis, CML is believed to
accumulate in elastic fibers in response to production of hydroxyl
radicals by ultraviolet rays, prompting active research into the
relationship between solar elastosis and AGEs.
While the relationship between f lecks and glycation is
unclear at present, the association between wrinkle formation
and glycation of collagen is considered to be one of the causes of
elastin glycation and the cross-linking of elastic fiber. Exposure
to ultraviolet rays induces the production of reactive oxygen
species (ROS)and thereby AGE formation. Prevention of
dermal aging and solar elastosis will therefore require inhibiting
not only glycation stress but also ROS production in response to
skin exposure to ultraviolet A radiation.

110

Glycation as a Risk Factor for Aging

Fig. 4. Receptors for AGEs.

further investigation. However, despite the increasingly apparent


importance of clarifying which AGE structures accumulate in
which tissue types, as AGE formation pathways and structure
are known to vary by pathological condition and tissue in which
they accumulate, no studies have yet thoroughly examined AGE
structures.
Clinical laboratories can measure AGE concentration
in blood using anti-AGE antibodies via an immunochemical
technique wherein serum samples are heated to 100 C for 15
minutes to inactivate the protease. However, heating biological
samples, as in foods, results artificial formation of AGEs such
as CML 25) and pentosidine 26) via Amadori rearrangement.
Development of an effective inhibitor of AGE formation in
anti-aging treatment will ultimately require (1) identifying the
structures of target AGEs, (2) analyzing the formation pathways
of target AGEs, and (3) developing a method of measuring
AGEs that does not involve the formation of artificial AGEs.

Recent research has focused on the relationship between


AGEs and regenerative medical techniques. AGEs have been
found to affect follicular growth, fertilization, embryonic
development, and pregnancy outcome 22), and AGE accumulation
is now believed to be a new index for rating ovarian dysfunction,
independent from age and day-3-FSH and therefore quite useful
for early diagnosis. In addition, reducing AGE accumulation may
represent a new method for treating infertility.
Several reports, including those by our group, have recently
demonstrated the presence of autoantibodies against AGEs
in human plasma. Shibayama et al. reported that the titer of
autoantibodies against CML in patients with diabetes was
significantly higher than in normal subjects 23). Similarly, Mera
et al. also found elevated levels of autoantibodies against several
AGE structures in plasma from healthy human subjects, with
titers of autoantibodies against N -(carboxyethyl)lysine (CEL)
particularly high compared with those of other AGE structures
such as CML and pentosidine (Fig. 5) 24). Further studies will be
needed to clarify the pathophysiological role of autoantibodies
against AGE structures and their potential to be used as
biological markers or therapeutic targets.

Conflict of interest statement:


This work was supported in part by Grants-in-Aid for
Scientific Research (No. 24300260 and 24650482 to Ryoji
Nagai) from the Ministry of Education, Science, Sports and
Cultures of Japan.

Future Research
As described above, in vivo research on AGEs is not limited
only to studies on diabetic complications but has extended into
various fields, consequently bringing to light points requiring
111

Glycation as a Risk Factor for Aging

Fig. 5. Levels of anti-AGE autoantibodies in healthy subjects.

112

Glycation as a Risk Factor for Aging

References
1) Nagai, R, Mori T, Yamamoto Y, et al: Significance of Advanced
Glycation End Products in Aging-Related Disease. Anti-Aging
Med. 7 (10) : 112-119, 2010
2) Mera K, Takeo K, Izumi M, et al: Effect of Reactive-Aldehydes
on the Modification and Dysfunction of Human Serum Albumin.
J. Pharm. Sci. 99 (3), 1614-1625, 2009
3) Suzuki H, Kurihara Y, Takeya N, et al: A role for macrophage
scavenger receptors in atherosclerosis and susceptibility to
infection. Nature 386(6633): 292-296, 1997
4) Ohgami N, Nagai R, Ikemoto M, et al: Cd36, a member of the
class b scavenger receptor family, as a receptor for advanced
glycation end products. J Biol Chem 276(5):3195-3202, 2001
5) Ohgami N, Nagai R, Miyazaki A, et al: Scavenger receptor class
B type I-mediated reverse cholesterol transport is inhibited by
advanced glycation end products. J Biol Chem 276(16):1334813355, 2001
6) Jono T, Miyazaki A, Nagai R, et al: Lectin-like oxidized low
density lipoprotein receptor-1 (LOX-1) serves as an endothelial
receptor for advanced glycation end products (AGE). FEBS Lett
511(1-3):170-174, 2002
7) Vlassara H, Li YM, Imani D, et al: Identification of galectin-3
as a high-affinity binding protein for advanced glycation end
products (AGE): a new member of the AGE-receptor complex.
Mol Med 1(6):634-646, 1995
8) Saito A, Nagai R, Tanu ma A, et al: Role of megalin in
endocytosis of advanced glycation end products: implications for
a novel protein binding to both megalin and advanced glycation
end products. J Am Soc Nephrol 14(5):1123-1131, 2003
9) Marsche G, Weigle B, Sattler W, et al: Soluble RAGE blocks
scavenger receptor CD36-mediated uptake of hypochloritemodified low-density lipoprotein. FASEB J 21(12):3075-3082,
2007
10) Yan SD, Chen X, Fu J, et al: RAGE and amyloid-beta peptide
neurotoxicity in Alzheimer's disease. Nature 382(6593):685-691,
1996
11) Hori O, Brett J, Slattery T, et al: The receptor for advanced
glycation end products (RAGE) is a cellular binding site for
amphoterin. Mediation of neurite outgrowth and co-expression
of rage and amphoterin in the developing nervous system. J Biol
Chem 270(43):25752-25761, 1995
12) Hofmann MA, Drury S, Fu C, et al: RAGE mediates a novel
proinflammatory axis: a central cell surface receptor for S100/
calgranulin polypeptides. Cell 97(7):889-901, 1999
13) Yamamoto Y, Harashima A, Saito H, et al: Septic shock is
associated with receptor for advanced glycation end products
ligation of LPS. J Immunol 186(5):3248-3257, 2011
14) He M, Kubo H, Morimoto K, et al: Receptor for advanced
glycation end products binds to phosphatidylserine and assists in
the clearance of apoptotic cells. EMBO Rep 12(4):358-364, 2011
15) Myint KM, Yamamoto Y, Doi T, et al: RAGE control of diabetic
nephropathy in a mouse model: effects of RAGE gene disruption
and administration of low-molecular weight heparin. Diabetes
55(9):2510-2522, 2006
16) Yonekura H, Yamamoto Y, Sakurai S, et al: Novel splice variants
of the receptor for advanced glycation end-products expressed
in human vascular endothelial cells and pericytes, and their
putative roles in diabetes-induced vascular injury. Biochem J
370(Pt 3):1097-1109, 2003

17) Win MT, Yamamoto Y, Munesue S, et al: Regulation of RAGE


for attenuating progression of diabetic vascular complications.
Exp Diabetes Res 2012:894605, 2012 Epub 2011 Oct 29
18) Mizutari K et al: Photo-enhanced modification of human skin
elastin in actinic elastosis by N -(carboxymethyl)lysine, one of
the glycoxidation products of the Maillard reaction. J. Invest.
Dermatol. 108, 797-802, 1997
19) Nagai R, Ikeda K, Higashi T, et al: Hydroxyl radical mediates
N -(carboxymethyl)lysine formation from Amadori product.
Biochem. Biophys. Res. Commun. 234, 167-172, 1997
20) Nagai R, Unno Y, Hayashi MC, et al: Peroxynitrite induces
formation of N -(carboxymethyl) lysine by the cleavage of
Amadori product and generation of glucosone and glyoxal
from glucose: novel pathways for protein modification by
peroxynitrite. Diabetes. 51, 2833-2839, 2002
21) Mera K, Nagai R, Haraguchi N, et al: Hypochlorous acid
generates N -(carboxymethyl)lysine from Amadori products.
Free Radic Res. 41, 713-718, 2007
22) Jinno M, Takeuchi M, Watanabe A, et al: Advanced glycation
e n d - p r o d u c t s a c c u m u l a t i o n c o m p r o m i s e s e m b r yo n i c
development and achievement of preg nancy by assisted
reproductive technology. Hum Reprod. 26(3), 604-610, 2011
23) Shibayama R, Araki N, Nagai R, et al: Autoantibody against N (carboxymethyl)lysine: an advanced glycation end product of the
Maillard reaction. Diabetes 48, 1842-1849, 1999
24) Mera K, Nagai R, Takeo K, et al: An autoantibody Against N (carboxyethyl)lysine (CEL): Possible Involvement in the Removal
of CEL-Modified Proteins by Macrophages. Biochem. Biophys.
Res. Commun. 407(2):420-425, 2011
25) Hayashi CM, Nagai R, Miyazaki K, et al: Conversion of Amadori
Products of the Maillard Reaction to N -(carboxymethyl)lysine
by Short-Term Heating: Possible Detection of Artifacts by
Immunohistochemistry. Lab Invest. 82, 795-808, 2002
26) Nakano M, Kubota M, Owada S, et al: The pentosidine
concentration in human blood specimens is affected by heating.
Amino Acids. 2011 Dec 4. [Epub ahead of print]

113

Das könnte Ihnen auch gefallen