You are on page 1of 10

Brain Research Reviews 23 Ž1997.

134–143

Short Review

Advanced glycation endproducts in ageing and Alzheimer’s disease


a,)
¨
Gerald Munch , Johannes Thome b, Paul Foley b, Reinhard Schinzel a , Peter Riederer b

a
Physiological Chemistry I, Theodor-BoÕeri-Institut (Biozentrum), Am Hubland, 97074 Wurzburg,
¨ Germany
b
¨
Clinical Neurochemistry, Department of Psychiatry, Fuchsleinstraße ¨
15, 97080 Wurzburg, Germany
Accepted 29 July 1996

Abstract

Accumulation of advanced glycation endproducts ŽAGE. in the brain is a feature of ageing and degeneration, especially in Alzheimer’s
disease ŽAD.. Increased AGE levels explain many of the neuropathological and biochemical features of AD such as extensive protein
crosslinking Žß-amyloid and MAP-t ., oxidative stress and neuronal cell death. Oxidative stress and AGEs initiate a positive feedback
loop, where normal age-related changes develop into a pathophysiological cascade. Combined intervention using antioxidants, metal
chelators, anti-inflammatory drugs and AGE-inhibitors may be a promising neuroprotective strategy.

Keywords: Advanced glycation end product; Neurodegeneration; Oxidative stress; Alzheimer’s disease; Plaque; Tangle; Neurotoxicity

Contents

1. Advanced glycation endproducts: chemistry . ...................................................... 135

2. Advanced glycation endproducts in ageing and degenerative diseases of the cardiovascular system . ......................... 136

3. Accumulation of advanced glycation endproducts in brain in ageing and in Alzheimer’s disease ........................... 136

4. Toxic effects of advanced glycation endproducts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137


4.1. Radical induced damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137
4.2. Induction of oxidative stress through binding to an advanced glycation endproduct receptor . . . . . . . . . . . . . . . . . . . . . . . . . . . 137
4.3. Inhibition of differentiation factor mediated neurite outgrowth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
4.4. Inhibition of protein turnover by advanced glycation endproducts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138

5. Factors for increased advanced glycation endproduct formation in Alzheimer’s disease . ............................... 138

6. Advanced glycation endproduct inhibitors: definition and proposed mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . ........ 139
6.1. Carnosine: a physiological advanced glycation endproduct inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ........ 139
6.2. Synthetic advanced glycation endproduct inhibitors: a new therapeutic opportunity for the treatment of Alzheimer’s disease? . ........ 139

7. Advanced glycation endproducts and their role in Alzheimer’s disease — a critical evaluation . ........................... 140

8. Conclusion ........................................................................ 141

Acknowledgements . ..................................................................... 141

References .......................................................................... 141

)
Corresponding author. Fax: q49 Ž931. 888-4150; E-mail: muench@biozentrum.uni-wuerzburg.de

0165-0173r97r$32.00 Copyright q 1997 Elsevier Science B.V. All rights reserved.


PII S 0 1 6 5 - 0 1 7 3 Ž 9 6 . 0 0 0 1 6 - 1
¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch 135

1. Advanced glycation endproducts: chemistry Table 1


Reactivities of different sugars Žwith ß-alanine as amino component. in
the Maillard reaction Žfrom w47x.
The amino groups of proteins, particularly the N-termi-
nal amino groups and side chains of lysine and arginine Sugar formation of radical formation
chromophores Žintensity of ESR
react non-enzymatically with monosaccharides, including Žbrowning. spectra.
glucose, fructose, hexose-phosphates, trioses and triose-
Glucose ŽC-6. 1 qq
phosphates. This posttranslational modification, termed Fruktose ŽC-6. 0.74 q
‘non-enzymatic glycosylation’, ‘glycation’ or ‘Maillard re- Xylose ŽC-5. 8.74 qq
action’, leads via reversible Schiff-base adducts to protein Mehylglyoxal ŽC-3. 654 not determined
bound Amadori products. Through subsequent oxidations Glyceraldehyde ŽC-3. 1967 qqq
and dehydrations, including free radical intermediates, a Glycolaldehyde ŽC-2. 2109 qqqq
broad range of heterogeneous fluorescent and yellow-brown
products with nitrogen- and oxygen-containing heterocy-
cles is formed, the so-called advanced glycation endprod-
ucts ŽAGEs. w4,5x. These latter reactions are accelerated by can be inhibited by metal chelators, emphasising the signif-
transition metals, such as copper and iron, which oxidise icance of transition metals for AGE-formation w76x. Among
the protein-bound Amadori-products. In this ‘glycoxida- the physiologically relevant sugars, glucose is the least
tion’ reaction not only protein-bound AGEs, but also solu- reactive, presumably the reason for its selection by evolu-
ble, highly reactive dicarbonyl products and oxygen free tion as the main biological energy carrier; the rank order of
radicals are formed. By an alternative route, transition reactivity for the other monosaccharides increases from
metals can oxidise the monosaccharides directly in solu- hexoses to trioses by several orders of magnitude ŽTable 1.
tion to form dicarbonyl products, which subsequently w47x. AGE formation is irreversible and causes protease-re-
crosslink proteins; this process is called ‘autooxidative sistant cross-linking of peptides and proteins, leading to
glycosylation’ w75x. The oxidation steps in both pathways protein deposition and amyloidosis ŽFig. 1..

Fig. 1. Chemistry of advanced glycation endproducts.


136 ¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch

2. Advanced glycation endproducts in ageing and de- animal brains, AGEs show a nuclear staining pattern w34x.
generative diseases of the cardiovascular system Pyramidal neurons selectively accumulate AGE-containing
vesicles in an age-dependent manner presumably in endo-
In the early 80’s, Monnier and Cerami, the pioneers of somes or lysosomes w30,35x. Interestingly, this type of
the ‘non-enzymatic glycosylation theory of ageing’ pro- neuron degenerates early in AD w35x. However, the AGE-
posed that the AGE-mediated crosslinking of long-lived antibodies used in the study by Li et al. were raised against
proteins contributes to the age-related decline in the func- an uncharacterised polylysine-glucose mixture and the re-
tion of cells and tissues in normal ageing w4,42,43,64x. sults should be interpreted rather cautiously.
Recent progress in the understanding of this process has The histological hallmarks of AD include widespread
confirmed that AGEs play a significant role in the evolu- neuronal cell death and the formation of amyloid plaques
tion of vascular complications in normal ageing, especially and neurofibrillary tangles. AGE modification and result-
in diabetes and renal failure. AGEs have been detected in a ing crosslinking of protein deposits has been shown to
variety of vascular wall, lipoprotein and lipid constituents, occur in both plaques and tangles which is not unexpected
where they lead to macroangiopathy, microangiopathy and since various long-lived and precipitated proteins become
amyloidosis. In particular, diseases such as atherosclerosis, modified by AGEs.
cataract and diabetic nephropathy, retinopathy and neu- In detail, increased extracellular AGE formation has
ropathy are suggested to be either caused or promoted by been demonstrated in amyloid plaques in different cortical
AGEs ŽTable 2. w4,52x. The detrimental effects of AGEs in areas w63,68x. In another immunohistochemical study vas-
pathological processes were originally attributed to their cular walls in amyloid angiopathy were not labelled by a
physicochemical properties including protein crosslinking monoclonal antibody, while primitive plaques, coronas of
but more recent studies increasingly emphasise the role of classic plaques and some glial cells in Alzheimer’s cortex
AGEs in cellular processes and signalling events, espe- were positive for AGEs w29x. These findings suggest that
cially in the oxidative stress response. In diabetes, acceler- AGE formation may occur in the early stages in plaque
ated AGE formation is caused primarily by a higher level formation in Alzheimer’s disease, but AGE-epitopes disap-
of plasma glucose and, intracellularly, by activation of the pear when the plaque ages or undergoes processing by
polyol pathway. In hemodialysis patients, similar cardio- astrocytes and microglia. There are only a few studies
vascular complications as in diabetic patients were ob- published so far showing that AGE formation not only
served, which led to the speculation that AGEs may be occurs subsequent to protein deposition, but actively accel-
also involved. These patients show an increased amount of erates their formation from soluble protein or peptide
AGEs with a molecular weight of about 2 kDa in their precursors. We and others could show that nucleation-de-
plasma, suggesting this may be caused by the inability of pendent polymerization of ß-amyloid peptide, the major
the dialysis cartridges to remove these low molecular component of plaques in patients with Alzheimer’s dis-
weight AGE-modified peptides w16,53,71x. However, the ease, is significantly accelerated by crosslinking through
factors responsible for the increased cerebral AGE levels AGEs in vitro w45,68x. This suggests, that AGE may
in Alzheimer’s disease ŽAD. are not yet fully understood. indeed represent a driving force in the acceleration of
ß-amyloid deposition and plaque formation. Numerous
studies have shown that susceptibility to AD is correlated
3. Accumulation of advanced glycation endproducts in with the dose of the e4 allele of apolipoprotein E ŽApo E.
brain in ageing and in Alzheimer’s disease w12,31x. Harrington and Colaco suggested that consequent
substitution of cysteines by arginines creates more glyca-
AGEs accumulate in pyramidal neurons in aged animals tion sites and hence facilitates the AGE-mediated cross-
Žhorse, calf, pig, and rat. and humans, but their intra- linking of Apo E to the insoluble deposits and hence
cellular distribution pattern is quite different between ani- accelerates plaque growth w20x.
mal and man. In the human pyramidal neuron, AGEs Neurofibrillary tangles ŽNFT. and neuropil threads are
exhibit a granular, perikaryonal distribution, whereas in further histological characteristics of Alzheimer’s disease
w78x. The rate of progression of Alzheimer’s disease-re-
lated neurofibrillary changes is unknown, but initial
Table 2 changes occur 50 years before the disease is diagnosed
Human degenerative diseases with a proposed involvement of AGEs
w2,3x. The major component of these tangles is the micro-
Failure of maintenance Major pathologies tubuli associated protein t ŽMAP-t ., which has been
in cells or tissues
shown to be subject to intracellular hyperglycation and
Neurones Dementias AGE formation w64x. Further, MAP-t is glycated at its
Retina, lens Blindness tubulin binding site, and MAP-t glycated in vitro is capa-
Insulin metabolism, signalling Complications of diabetes
Blood vessels Cardiovascular diseases
ble of inducing oxidative stress w33,77x. The protein con-
Glomeruli Renal failure stituents of NFT are resistant to proteolytic removal, possi-
bly as a result of extensive cross-links w10x. Although it is
¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch 137

reasonable to accept the idea of AGE-modification of glycated protein w46x. This process commences with the
extracellular tangles, it is not clear to what extent glycation production of superoxide radicals by transition metal-cata-
is involved in the first steps of intracellular tangle forma- lysed autoxidation of the sugars and protein bound Amadori
tion, especially when one considers the established cross- products, followed by dismutation of superoxide to hydro-
linking and aggregation mechanism such as formation of gen peroxide and the generation of lethal hydroxyl radicals
specific disulfide bridges and hyperphosphorylation w36,60x by the Fenton reaction. This leads to a site-specific attack
or formation of core fragments of MAP-t w50x. on the protein with consequent protein damage and lipid
In summary, among the many factors proposed to be peroxidation w64x.
involved in the etiology or progression of Alzheimer’s AGE can also produce oxygen free radicals through an
disease, AGEs are a relatively new and interesting ap- indirect, immune system mediated process. Interaction of
proach to unravel the mysteries of the etiopathogenesis of AGE-modified proteins with microglia in an acute phase
this multifactorial disease. However, one has to be careful reaction can lead to a ‘respiratory burst’. This radical
not to overestimate their role in the disease process as long challenge has been shown to cause ‘bystander-lysis’ of
as only circumstantial evidence is presented that they are neighbouring neurons ŽFig. 2. w41x. AGEs have been shown
active promotors of the progression, not only simply a to induce inflammatory processes in regions other than the
secondary epiphenomenon in Alzheimer’s disease w56x. brain such as activation of peripheral macrophages w18x
and chemotaxis of mononuclear phagocytes w57x. AGEs
also induce the release of cytokines such as interleukins 1
4. Toxic effects of advanced glycation endproducts and 6, as well as of TNF-a w70x. Interestingly, IL-6
immunoreactivity has previously been demonstrated in
AGEs have been shown to be more than a harmless plaques in Alzheimer’s disease, and elevated IL-6 concen-
post-translational protein modification; various pathophysi- trations have been measured in brains of AD patients w26x.
ological effects have been found at the cellular and molec- This concurs with the finding that an autodestructive pro-
ular level. Among the proposed mechanisms of AGE-in- cess, involving overactive astroglia and microglia, occurs
duced damage are the following four. at the characteristic lesions in Alzheimer disease w39,40,73x.

4.1. Radical induced damage


4.2. Induction of oxidatiÕe stress through binding to an
Associated with the oxidation of sugars and Amadori adÕanced glycation endproduct receptor
products is the formation of oxygen free radicals. Protein
glycation increases the rate of free radical production at Binding of AGEs to specific receptors can generate
physiological pH nearly 50-fold compared with non- oxidative stress, as measured chemically by the production

Fig. 2. Activation of microgliarmacrophages by AGE-modified protein Že.g., ß-amyloid. deposits and resulting cytotoxicity.
138 ¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch

of thiobarbituric acid-reactive substances. It might be pos- dation, which would be expected to shift the metabolic
sible, that this occurs through an oxidative signalling path- balance in the direction of deposition rather than degrada-
way or through internalisation of AGEs by one of the tion. Proteins modified by sugars more reactive than glu-
many characterized AGE-receptors. Beside one recently cose, such as fructose, are more resistant to proteolysis,
described receptor for AGEs, RAGE w57–59,74x and other suggesting that proteolysis is a complex process dependent
receptors and receptor complexes with binding specificity on the various protein-bound moieties generated at differ-
for AGEs w56,69,72x, it is known that the macrophage ent stages of the Maillard reaction w65x. Metabolic transit
scavenger receptor ŽMSR. mediates the endocytic uptake experiments have shown, that the AGE moieties them-
and degradation of AGE proteins w1x. In the brain, this selves can only be degraded by the bacteria found in the
receptor is expressed on microglia, but not on astrocytes, digestive tract, but not directly by the host w15x. We have
neurons, or vessel-associated structures. In Alzheimer dis- shown that the activity of certain proteases Žcathepsins.
ease, there is strong expression of the scavenger receptor and the total cellular protein turnover are attenuated in
in association with senile plaques w11x. cells exposed to synthetic AGEs w61x.
Oxidative stress in AD it is not only evidenced by
chemical markers w17x but also by the activation of the
antioxidative defence system, such as the activation of the
transcription factor NFk B and the upregulation of heme 5. Factors for increased advanced glycation endproduct
oxygenase. These AGE-mediated effects can be blocked formation in Alzheimer’s disease
by addition of antioxidants, such as probucol, or of anti-
bodies to the AGE-receptor w74x. In AD tissue, accumu- AGEs play an important role in the evolution of vascu-
lated paired helical filament-t in neurons is also subject to lar complications in normal ageing as discussed in Section
non-enzymatic glycation and AGE formation, and these 2. AGEs in diabetes are generally associated with a higher
neurons also exhibit evidence of oxidative stress, including level of plasma glucose and in renal failurerhemodialysis
the production of malondialdehyde and the heme oxyge- by the inability of the dialysis cartridges to remove AGE-
nase 1 antigen. In cell culture experiments, PHF-t isolated modified peptides w4,16,71x. However, many additional
from post mortem tissue and recombinant AGE-t each factors including the involvement of C-3 and C-2 sugars
generate oxygen free radicals, thereby not only activating and fragmentation products, even oxidised ascorbate w48x,
transcription via NFk B, but also increasing APP levels as well as transition metals and oxidative stress contribute
and inducing the release of the characteristic 4-kDa ß- to AGE formation in different tissues. Although thor-
amyloid-peptides w58,77x. oughly investigated for about nearly 20 years, the chem-
istry and biochemistry of glycoxidation are very complex
4.3. Inhibition of differentiation factor mediated neurite and many critical issues, such as tissue and protein specific
outgrowth glycemic thresholds w49x, still have to be resolved. In
analogy to diabetes, many of the factors mentioned above
may explain the elevated level of AGEs and AGE
One putative receptor for advanced glycation end prod-
crosslinked proteins in the brain tissue of AD patients. In
ucts ŽRAGE., a member of the immunoglobulin superfam-
detail, the following three specific changes in AD may
ily, mediates interactions of AGE-modified proteins with
contribute to this process:
endothelium and other cell types. AGE binding to this type
Ži. an intracellular increase in particular AGE reactive
of receptors not only induces an oxidative stress response
sugars as the consequence of a disturbed glucose
but may also antagonise the binding of other specific
metabolism w24,25x;
physiological ligands. RAGE has physiologically relevant
Žii. an increase in unchelated transition metals such as
ligands distinct from AGEs, including amphoterin, which
copper and iron, acceleration of the oxidation of glycated
mediates neurite outgrowth in the developing CNS. A
proteins and subsequent increase in highly reactive glycox-
non-specific interference with such growth or differentia-
idation products w9,75x,
tion factors may also be a pathophysiological effect of
Žiii. depletion of the antiglycation substance pool, for
AGEs w23x.
example the histidine dipeptides including carnosine and
anserine w21,22x.
4.4. Inhibition of protein turnoÕer by adÕanced glycation Interestingly, the increased level of cerebral AGEs in
endproducts AD patients is not reflected by an increased AGE level in
plasma w67x. This is not an unexpected finding as an
It has been speculated that a general proteolytic imbal- increased AGE plasma level would lead to cardiovascular
ance in the AD brain contributes to the accumulation of complications and the dementia would be classified as a
amyloid plaques and neurofibrillary tangles w62x. AGE- vascular type dementia. In summary, cerebral AGE accu-
modification not only contributes to protein crosslinking, it mulation in AD is probably a highly selective, brain
also decreases the susceptibility to proteolysis and degra- specific event.
¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch 139

6. Advanced glycation endproduct inhibitors: definition


and proposed mechanism

An AGE-inhibitor is usually defined as a substance,


which inhibits the covalent crosslinking of proteins and
peptides by sugars or sugar derived oxidation products. It
contains a nucleophilic amino group which competes with
lysines side chains of proteins for reactive carbonyl or
dicarbonyls groups formed on proteins and in solution.
Since the crucial step of AGE crosslinking depends on the
presence of transition metals for the formation of glycoxi-
dation products and radicals, metal chelators and radical
scavengers, especially superoxide dismutase mimetics,
could also be regarded as AGE-inhibitors in a broader Fig. 4. Correlation of skeletal muscle carnosine concentration with lifes-
sense of the definition. Several natural and synthetic com- pan in different animals.
pounds ŽFig. 3. have been shown to be inhibitors of
AGE-formation in vitro and in vivo w6,19x.
Carnosine inhibits glycation of acetyl-Lys-His-amide
6.1. Carnosine: a physiological adÕanced glycation end- Žthe preferred glycation site on proteins. and protects
product inhibitor model proteins Že.g., a-crystalline, superoxide dismutase
and catalase. against glycation and AGE cross-linking
The dipeptide carnosine Žß-alanyl-L-histidine. is widely w21,22x. Mammals thus possess strategies using physio-
distributed in mammalian tissues, including muscle and logical AGE-inhibitors such as carnosine which minimise
brain with concentrations of up to 20mM w28,51x. It is glycation and AGE-formation. Dysfunction of the physio-
synthesised by tissue carnosine synthetase from its compo- logical AGE-defence and -clearance mechanisms could
nent amino acids, and degraded by carnosinase, predomi- underlie a variety of degenerative diseases, including
nantly in plasma. Various biological functions of carnosine Alzheimer’s disease. On the other hand, supplementing
have been demonstrated, including its role as a antioxidant, naturally occurring anti-glycation molecules by synthetic
metal-chelator, SOD mimetic and radical scavenger w55x. AGE-inhibitors, especially in tissues with low concentra-
One of the most prominent effects of carnosine is its tions of natural compounds could be an effective therapeu-
anti-ageing properties including as life span extension and tic strategy for Alzheimer’s disease.
the prevention of the appearance of the usual signs of
late-passage senescence Žinability to line up in parallel 6.2. Synthetic adÕanced glycation endproduct inhibitors: a
arrays in a confluent monolayer. in fibroblasts cells w38x. new therapeutic opportunity for the treatment of
Interestingly, carnosine concentrations in skeletal muscle Alzheimer’s disease?
correlates with the life-span of the corresponding animal
ŽFig. 4. w7,37,55x. In addition, histidine dipeptide levels Aminoguanidine was the ‘pioneering’ AGE-inhibitor,
Žcarnosine, anserine, etc.. decrease in skeletal and cardiac which was first tested in in vitro crosslinking experiments,
muscles in ageing rats w28x. Although the data in both then shown in various animal models to attenuate the
studies seem reliable, in general, claims that certain drugs AGE-mediated vascular complications of diabetes w6,19x.
or physiological substances which decline with age, corre- Tenilsetam , Ž q . -3- Ž 2-thienyl . -2-piperazinone, a
late with life span or can even be used for its extension, cognition-enhancing and a purportedly anti-dementia drug
should be analyzed very critically. was also shown to be an effective AGE inhibitor, although
its mechanism is yet not fully understood. Tenilsetam
reacts with sugars and glycated proteins and acts as an
inhibitor of AGE-induced amino acid and protein cross-
linking in vitro w44x. Tenilsetam, aminoguanidine and
carnosine significantly inhibit nucleation-dependent poly-
merisation of ß-amyloid peptide with similar efficacy w45x.
The positive effects of AGE-inhibitors with respect to the
reduction of ß-amyloid toxicity may be attributable to
formation of ‘masked’, less toxic AGE peptide aggregates.
AGE-inhibitors could also shift the metabolic AGE bal-
ance in the direction of degradation and clearance rather
Fig. 3. Chemical structures of the AGE-inhibitors aminoguanidine, carno- than accumulation. AGE-inhibitors could also modify the
sine and tenilsetam. structure of AGEs in a way which inhibits their binding to
140 ¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch

and recognition by AGE receptors. These mechanisms carnosine and aminoguanidine might be limited to the
might subsequently lead to a diminished inflammatory cardiovascular system, if they are not actively transported
response and decreased oxidative stress ŽFig. 5.. However, across the blood brain barrier.
these are quite speculative hypotheses and need substantial Despite some circumstantial evidence that the beneficial
investigations at the molecular and cellular level before effect of tenilsetam in AD patients supports the involve-
incorporating them into the etiopathogenesis of AD w66x. ment of AGEs in pathological processes in the late stages
However, some pharmacological support for these hy- of AD, a direct attenuation of cytotoxic AGE effects by
potheses comes from the first clinical trial with an AGE- AGE-inhibitors has to be shown at least before proposing
inhibitor, tenilsetam, in AD patients. This drug signifi- this as a valid approach for the treatment of AD patients.
cantly improved many clinical and psychometric scores
including shortening of P300 latencies in the acoustic
evoked potential, global clinical impression, Sandoz clini- 7. Advanced glycation endproducts and their role in
cal assessment geriatric scale, Folstein minimental state Alzheimer’s disease — a critical evaluation
scale and shopping list w13,27x. Interestingly, the improve-
ment had not reached a plateau when the study was ended Alzheimer’s disease, the most common dementing dis-
after three months. Although a direct cholinergic mecha- order of late life, is a major cause of disability and death in
nism was initially assumed for tenilsetam, there was no the elderly. Neurobiological, genetic, and molecular stud-
evidence for muscarinic acetylcholine receptor binding or ies have defined the vulnerable neural systems, including
acetylcholine re-uptake inhibition. No evidence for an abnormalities in cytoskeletal proteins in neurons, the biol-
alternative neuroprotective mechanism such as free radical ogy of the beta-amyloid precursor protein and its prote-
scavenging was found. However, the slow appearance of olytic cleavage product, beta-amyloid w12,14,54x. The mul-
clinical improvement suggests that the efficacy in tifactorial puzzle of the disease includes most likely a
Alzheimer’s patients may be indeed due to interference combination of age-related degenerative changes with ad-
with post-translational modification of proteins. Tenilse- ditional genetic predispositions and environmental factors
tam, a non-charged molecule, crosses the blood brain determining the vulnerability of the individual. Although
barrier, but the effects of the charged AGE-inhibitors there might be various different critical triggering events in

Fig. 5. Suggested role of AGEs in the pathogenesis of Alzheimer’s disease and points of possible therapeutic intervention with AGE-inhibitors.
¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch 141

the early stages of the disease, they seem to converge on a Acknowledgements


few characteristic final pathways in the late stages of the
disease. It seems reasonable to believe, that not the initial We thank A. Hipkiss, J. Michaelis, R. Holliday, U.
events, but the later steps most likely offer the most Schindler, A.M. Cunningham, M. Atkinson and J. Shine
promising pharmalogical approaches for slowing down the for valuable and critical discussions and the ‘Claussen-
progression of the disease. Stiftung’ and the ‘Hirnliga e.V.’ for funding.
However, before a specific treatment by AGE-inhibitors
can be proposed for AD, it has to be proven beyond doubt
that AGEs are more than just a harmless post-transational References
protein modification, but an irreversible promoter of cross-
linking and maybe even a cytotoxic substance. It has been w1x Araki, N., Higashi, T. Mori, T., Shibayama, R., Kawabe, Y.,
shown, that AGEs accelerate the formation and inhibit the Kodama, T., Takahashi, K., Shichiri, M. and Horiuchi, S.,
removal of AGE-modified protein deposits w5,45,68x. If Macrophage scavenge receptor mediates the endocytic uptake and
degradation of advanced glycation end products of the Maillard
these proteins, e.g., the ß-amyloid peptide, are toxic them- reaction, Eur. J. Biochem., 230 Ž1995. 408–415.
selves w32x, this might be enough to suggest a ‘supporting w2x Braak, H. and Braak, E., Staging of Alzheimer’s disease-related
role’ of AGEs in the pathogenesis of AD. However, it is neurofibrillary changes, Neurobiol. Aging, 16 Ž1995. 271–278.
necessary to investigate if AGEs themselves exert a direct w3x Braak, E., Braak, H. and Mandelkow, E.M., A sequence of cyto-
toxicity to cells. Studies about the indirect neurotoxicity of skeleton changes related to the formation of neurofibrillary tangles
and neuropil threads, Acta Neuropathol., 87 Ž1994. 554–567.
AGEs by activation of microglia and subsequent radical w4x Brownlee, M., Advanced protein glycosylation in diabetes and ag-
and cytokine release seem to be quite sound, but it is not ing, Annu. ReÕ. Med., 46 Ž1995. 223–234.
clear, if this event in AD occurs before the neurons have w5x Buccala, R. and Cerami, A., Advanced glycosylation: chemistry,
been already irreversibly damaged. biology, and implications for diabetes and aging, AdÕ. Pharmacol.,
At this point of time, it seems quite speculative and 23 Ž1992. 1–34.
w6x Cameron, N.E. and Cotter, M.A., Potential therapeutic approaches to
premature to propose a specific treatment of AD with the treatment or prevention of diabetic neuropathy: evidence from
AGE-inhibitors as a rational approach to the treatment of experimental studies, Diabetes Med., 10 Ž1993. 593–605.
the disease, as long as a direct attenuation of the direct or w7x Carnegie, P.R., Ilic. M.Z., Etheridge, M.O. and Collins, M.G.,
indirect cytotoxic effects of AGEs in cell culture or in an Improved high performance liquid chromatograph method for analy-
animal model by AGE-inhibitors have not been shown. sis of histidine dipeptides anserine, carnosine and balenin present in
fresh meat, J. Chromatogr., 211 Ž1983. 153–157.
However, one has to keep in mind, that there is no w8x Chun, M. R. and Mayeux, R., Alzheimer’s disease, Curr. Opin.
promising alternative treatment for AD avaliable, since the Neurol., 7 Ž1994. 299–304.
beneficial effects of cholinergic substitution therapy has w9x Connor, J.R., Tucker, P., Johnson, M. and Snyder, B., Ceruloplas-
definitely fallen short of expectations w8x. min levels in the human superior temporal gyrus in aging and
Alzheimer’s disease, Neurosci. Lett., 159 Ž1994. 88–90.
w10x Cras, P., Smith, M.A., Richey, P.L., Siedlak, S.L., Mulvihill, P. and
Perry, G., Extracellular neurofibrillary tangles reflect neuronal loss
8. Conclusion and provide further evidence of extensive protein cross-linking in
Alzheimer disease, Acta Neuropathol., 89 Ž1995. 291–295.
Many of the degenerative changes described in AD, w11x Christie, R.H., Freeman, M. and Hyman, B.T., Expression of the
such as increased oxidative stress and formation of AGEs macrophage scavenger receptor, a multifunctional lipoprotein recep-
tor, in microglia associated with senile plaques in Alzheimer’s
resemble processes seen in other diseases including late disease, Am. J. Pathol., 148 Ž1996. 399–403.
complications in diabetes and long term hemodialysis. The ¨
w12x Czech, C., Forstl, H., Hentschel, F., Monning, U., Besthorn, C.,
‘glycation theory of ageing’ as the underlying common Geiger-Kabisch, C., Sattel, H., Masters, C. and Beyreuther K,
principle of degeneration unites most of the neuropatholog- Apolipoprotein e-4 gene dose in clinically diagnosed Alzheimer’s
ical and biochemical findings in AD to a general picture. disease: prevalence, plasma cholesterol levels and cerebrovascular
change, Eur. Arch. Psychiatry Clin. Neurosci., 243 Ž1995. 291–292.
AGE formation explains accelerated protein crosslinking w13x Dierks, T., Maurer, K. and Ihl, R., Influence of tenilsetam on
with ß-amyloid and MAP-t, increased oxidative stress, and AEP-300 in Alzheimer’s disease, J. Neural Transm., wP-D-Sectx 1
direct as well as indirect immune system mediated induc- Ž1989. 49.
tion of neuronal cell death w20,64x. In particular, oxidative ¨
w14x Eckert, A., Forstl, H., Hartmann, H., Czech, C., Monning, U.,
stress and AGEs amplify each others’ effects and produce ¨
Beyreuther, K. and Muller, W.E., The amplifying effect of beta-
amyloid on cellular calcium signalling is reduced in Alzheimer’s
a positive feedback loop of degeneration. If this vicious disease, NeuroReport, 6 Ž1995. 1199–1202.
circle cannot be physiologically managed, normal age-re- w15x Finot, P.A. and Furniss, D.E., Metabolic transit and toxicity of
lated alterations become a pathological cascade resulting in Maillard reaction products, Prog. Clin. Biol. Res., 304 Ž1989.
irreversible damage. Pharmacological intervention using 343–358.
w16x Friedlander, M.A., Wu, Y.C., Schulak, J.A., Monnier, V.M. and
antioxidants, metal chelators and AGE-inhibitors may be a
Hricik, D.E., Influence of dialysis modality on plasma and tissue
promising combined approach for minimising AGE forma- concentrations of pentosidine in patients with end-stage renal dis-
tion in ageing and degeneration in general and, especially, ease, Am. J. Kidney. Dis., 25 Ž1995. 445–451.
the resulting pathological effects in Alzheimer’s disease. ¨ M.E., Kunig,
w17x Gotz, ¨ G., Riederer, P. and Youdim, M.B., Oxidative
142 ¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch

stress: free radical production in neural degeneration, Pharmacol. quadriceps muscle of healthy humans, Eur. J. Appl. Physiol., 64
Ther., 63 Ž1994. 37–122. Ž1992. 47–50.
w18x Guzdek, A. and Stalinska, K, Glycated proteins as inducers of acute w38x McFarland, G.M. and Holliday, R., Retardation of the senescence of
phase cytokines, Fol. Histochem. Cytobiol., 30 Ž1992. 167–169. cultured human diploid fibroblasts by carnosine, Exp. Cell Res., 212
w19x Hammes, H.P., Brownlee, M., Edelstein, D., Saleck, M., Martin, S. Ž1994. 167–175.
and Federlin, K, Aminoguanidine inhibits the development of accel- w39x McGeer, P.L., Rogers, J. and McGeer, E.G., Neuroimmune mecha-
erated diabetic retinopathy in the spontaneous hypertensive rat, nisms in Alzheimer disease pathogenesis, Alzheimer Dis. Assoc.
Diabetologia, 37 Ž1994. 32–35. Disord., 8 Ž1994. 149–158.
w20x Harrington, C.R. and Colaco, C.A., Alzheimer’s disease. A glyca- w40x McGeer, P.L., Walker, D.G., Akiyama, H., Yasuhara, O. and
tion connection, Nature, 370 Ž1994. 247–248. McGeer, E.G., Involvement of microglia in Alzheimer’s disease,
w21x Hipkiss, A.R., Michaelis, J., Syrris, P., Kumar, S. and Lam, Y., Neuropathol. Appl. Neurobiol., 20 Ž1994. 191–192.
Carnosine protects proteins against in vitro glycation and cross-lin- w41x McMillian, M., Kong, L.Y., Sawin, S.M., Wilson, B., Das, K.,
king, Biochem. Soc. Trans., 2 Ž1994. 399. Hudson, P., Hong, J.S. and Bing, G., Selective killing of cholinergic
w22x Hipkiss, A.R., Michaelis, J. and Syrris, P., Non-enzymatic glyco- neurons by microglial activation in basal forebrain mixed
sylation of the dipeptide L-carnosine, a potential anti-protein-cross- neuronalrglial cultures, Biochem. Biophys. Res. Commun., 215
linking agent, FEBS Lett., 371 Ž1995. 81–85. Ž1995. 572–577.
w23x Hori, O., Brett, J., Slattery, T., Cao, R., Zhang, J., Chen, J.X., w42x Monnier, V.M., Towards a Maillard reaction theory of aging, Prog.
Nagashima, M., Lundh, E.R., Vijay S, Nitecki, D. and Stern, D.; Clin. Biol. Res., 304 Ž1989. 1–22.
The receptor for advanced glycation end products ŽRAGE. is a w43x Monnier, V.M. and Cerami, A., Nonenzymatic browning in vivo:
cellular binding site for amphoterin. Mediation of neurite outgrowth possible process for aging of long-lived proteins, Science, 211
and co-expression of rage and amphoterin in the developing nervous Ž1981. 491–493.
system, J. Biol. Chem., 270 Ž1995. 25752–25761. w44x ¨
Munch, G., Taneli, Y., Schraven, E., Schindler, U., Schinzel, R.,
w24x Hoyer, S., Neurodegeneration, Alzheimer’s disease, and beta-amyloid Palm, D. and Riederer, P., The cognition-enhancing drug Tenilsetam
toxicity, Life Sci., 55 Ž1994. 1977–1983. is an inhibitor of protein crosslinking by advanced glycosylation, J.
w25x Hoyer, S., Age-related changes in cerebral oxidative metabolism. Neural Transm., wP-D-Sectx 8 Ž1994. 193–208.
Implications for drug therapy, Drugs Aging, 6 Ž1995. 210–218 w45x ¨
Munch, G., Mayer, S., Michaelis, J., Hipkiss, A.R., Schinzel, R.,
w26x ¨ M., Strauss, S., Volk, B., Berger, M. and Bauer, J., Inter-
Hull, Riederer, P. and Cunningham, A.M., Influence of advanced glyco-
leukin-6 is present in early stages of plaque formation and is sylation endproducts and AGE-inhibitors on nucleation-dependent
restricted to the brains of Alzheimer’s disease patients, Acta Neu- polymerization of ß-amyloid peptide, Biochem. Biophys. Acta, Ž1996.
ropathol., 89 Ž1995. 544–551. in press.
w27x Ihl, R., Perisic, I., Maurer, K. and Dierks, T., Effect of 3 months w46x Mullarkey, C.J., Edelstein, D. and Brownlee, M., Free radical
treatment with tenilsetam in patients suffering from dementia of generation by early glycation products: a mechanism for accelerated
Alzheimer type ŽDAT., J. Neural Transm., wP-D Sectx 1 Ž1989. atherogenesis in diabetes, Biochem. Biophys. Res. Commun., 173
84–85. Ž1990. 932–939.
w28x Johnson, P. and Hammer, J.L., Histidine dipeptide levels in ageing w47x Namiki, M., Chemistry of Maillard reactions: recent studies on the
and hypertensive rat skeletal and cardiac muscles, Comp. Biochem. browning reaction mechanism and the development of antioxidants
Physiol., wBx 103 Ž1992. 981–984. and mutagens, AdÕ. Food Res., 32 Ž1988. 115–184.
w29x Kimura, T., Takamatsu, J and Araki, K., Are advanced glycation w48x Nagaraj, R.H.and Monnier, V.M., Isolation and characterization of a
end-products associated with amyloidosis in Alzheimer’s disease? blue fluorophore from human eye lens crystallins: in vitro formation
NeuroReport, 6 Ž1995. 866–868. from Maillard reaction with ascorbate and ribose, Biochim. Biophys.
w30x Kimura, T., Takamatzsu, J., Ikeda, K., Kondo, A., Miyakawa, T. Acta, 1116 Ž1992. 34–42
and Horiuchi, S., Accumulation of advanced glycation end products w49x Nagaraj, R.H, Kern, T.S., Sell, D.R., Fogarty, J., Engerman, R.L.
of the maillard reaction with age in human hippocampal neurons, and Monnier, V.M., Evidence for a glycemic threshold for the
Neurosci. Lett., 208 Ž1996. 53–56. formation of pentosidine in diabetic dog lens but not in collagen,
w31x ¨
Kurz, A., Lautenschlager, N., Haupt, M., Zimmer, R., von Thulen, Diabetes, 45 Ž1996. 587–594.
¨
B., Altland, K., Lauter, H. and Muller, U., Das Apolipoprotein w50x Novak, M., Kabat, J. and Wischik, C. M., Molecular characteriza-
E-e4-Allel ist ein Risikofaktor fur ¨ die Alzheimer-Krankheit mit tion of the minimal protease resistant t unit of the Alzheimer’s
¨
fruhem ¨
und spatem Beginn, NerÕenarzt, 65 Ž1994. 774–779 disease paired helical filament, EMBO J., 12 Ž1993. 365–370
w32x Lambert, M.P., Stevens, G., Sabo, S., Barber, K., Wang, G., Wade, w51x O’Dowd, J.J., Cairns, M.T., Trainor, M., Robins, D.J. and Miller,
W., Krafft, G., Snyder, S., Holzman, T.F. and Klein, W.L., D.J., Analysis of carnosine, homocarnosine, and other histidyl
BetarA4-evoked degeneration of differentiated SH-SY5Y human derivatives in rat brain, J. Neurochem., 55 Ž1990. 446–452.
neuroblastoma cells, J. Neurosci. Res., 39 Ž1994. 377–385. w52x Palinski, W., Koschinsky, T., Butler, S.W., Miller, E., Vlassara, H.,
w33x Ledesma, M.D., Bonay, P. and Avila, J., T protein from Alzheimer’s Cerami, A. and Witztum, J.L., Immunological evidence for the
disease patients is glycated at its tubulin-binding domain, J. Neu- presence of advanced glycosylation end products in atherosclerotic
rochem., 65 Ž1995. 1658–1664. lesions of euglycemic rabbits, Arterioscler. Thromb. Vasc. Biol., 15
w34x Li J.J., Voisin, D., Quiquerez, A.L. and Bouras C., Differential Ž1995. 571–582
expression of advanced glycation end-products in neurons of differ- w53x Papanastasiou, P., Grass, L., Rodela, H., Patrikarea, A., Oreopoulos,
ent species, Brain Res., 641 Ž1994. 285–288. D. and Diamandis, E.P., Immunological quantification of advanced
w35x Li, J.J., Surini, M., Catsicas, S., Kawashima, E. and Bouras, C., glycosylation end-products in the serum of patients on hemodialysis
Age-dependent accumulation of advanced glycosylation end prod- or CAPD, Kidney Int., 46 Ž1994. 216–222.
ucts in human neurons, Neurobiol. Aging 16 Ž1995. 69–76. w54x Price, D.L. and Sisodia, S.S., Cellular and molecular biology of
w36x Mandelkow, E.M., Schweers, O., Drewes, G., Biernat, J., Gustke, Alzheimer’s disease and animal models, Annu. ReÕ. Med., 45
N., Trinczek, B. and Mandelkow, E., Structure, microtubule interac- Ž1994. 435–446.
tions, and phosphorylation of t protein, Ann. NY Acad. Sci., 777 w55x Quinn, P.J., Boldyrev, A.A. and Formazuyk, V.E., Carnosine: its
Ž1996. 96–106. properties, function and potential therapeutic applications, Mol.
w37x Mannion, A.F., Jakeman, P.M., Dummett, M., Harris, R.C. and Aspects Med., 13 Ž1992. 379–444.
William, P.L. Ž1992. Carnosine and anserine concentrations in the w56x Shaw, S.M. and Crabbe, M.J., Characterisation of a putative scav-
¨ et al.r Brain Research ReÕiews 23 (1997) 134–143
G. Munch 143

enger receptor for advanced glycosylation end-products, Biochem. w68x Vitek, M. P., Bhattacharya, K., Glendening, J. M., Stopa, E.,
Soc. Trans., 22 Ž1994. 82S. Vlassara, H., Bucala, R., Manogue, K. and Cerami, A., Advanced
w57x Schmidt, A.M., Yan, S.D., Brett, J., Mora, R., Nowygrod, R. and glycation end products contribute to amyloidosis in Alzheimer dis-
Stern D., Regulation of human mononuclear phagocyte migration by ease, Proc. Natl. Acad. Sci. USA, 91 Ž1994. 4766–4770.
cell surface-binding proteins for advanced glycation end products, J. w69x Vlassara, H., Brownlee, M. and Cerami, A., Novel macrophage
Clin. InÕest., 91 Ž1993. 2155–2168. receptor for glucose-modified proteins is distinct from previously
w58x Schmidt, A.M., Hori, O., Brett, J., Yan, S.D., Wautier, J.L. and described scavenger receptors, J. Exp. Med., 164 Ž1986. 1301–1309.
Stern, D., Cellular receptors for advanced glycation end products. w70x Vlassara, H., Brownlee, M., Manogue, K., Dinarello, C. and
Implications for induction of oxidant stress and cellular dysfunction Pasagian, A., CachectinrTNF and IL-1 induced by glucose-mod-
in the pathogenesis of vascular lesions, Arterioscler. Thromb., 14 ified proteins: role in normal tissue remodeling, Science, 240 Ž1988.
Ž1994. 1521–1528. 1546.
w59x Schmidt, A.M., Hori, O., Chen, J.X., Li, J.F., Crandall, J., Zhang, J., w71x Vlassara, H., Serum advanced glycosylation end products: a new
Cao, R., Yan, S.D., Brett, J and Stern, D., Advanced glycation class of uremic toxins? Blood Purif., 12 Ž1994. 54–59.
endproducts interacting with their endothelial receptor induce ex- w72x Vlassara, H., Li, Y.M., Imani, F., Wojciechowicz, D., Yang, Z., Liu,
pression of vascular cell adhesion molecule-1 ŽVCAM-1. in cultured F.T. and Cerami, A., Identification of galectin-3 as a high-affinity
human endothelial cells and in mice. A potential mechanism for the binding protein for advanced glycation end products ŽAGE.: a new
accelerated vasculopathy of diabetes, J. Clin. InÕest., 96 Ž1995. member of the AGE-receptor complex, Mol. Med., 1 Ž1995. 634–
1395–1403. 646.
w60x Schweers, O., Mandelkow, E. M., Biernat, J. and Mandelkow, E., w73x Walker, D.G. and McGeer, P.L., Complement gene expression in
Oxidation of cysteine-322 in the repeat domain of microtubule-asso- human brain: comparison between normal and Alzheimer’s disease
ciated protein t controls the in vitro assembly of paired helical cases, Mol. Brain Res., 14 Ž1992. 109–116.
filaments, Proc. Natl. Acad. Sci. USA, 92 Ž1995. 8463–8467. w74x Wautier, J.L., Wautier, M.P., Schmidt, A.M., Anderson, G.M., Hori,
w61x Sebetkova, K., Ling, H., Schinzel, R., Simm, A., Vamvakas, S., O., Zoukourian, C., Capron, L., Chappey, O., Yan, S.D. and Brett,
Schenk, O., Munch, ¨ G., and Heidland, A., Advanced glycation J., Advanced glycation end products ŽAGEs. on the surface of
endproducts ŽAGEs. impair protein degradation in LLC-PK1 cells, diabetic erythrocytes bind to the vessel wall via a specific receptor
Ž1996. Žsubmitted.. inducing oxidant stress in the vasculature: a link between surface-as-
w62x Smith, M.A. and Perry, G., Alzheimer disease: an imbalance of sociated AGEs and diabetic complications, Proc. Natl. Acad. Sci.
proteolytic regulation? Med. Hypotheses, 42 Ž1994. 277–279. USA, 91 Ž1994. 7742–7746.
w63x Smith, M.A., Taneda, S., Richey, P.L., Miyata, S., Yan, S,D., Stern, w75x Wells-Knecht, K.J., Zyzak, D.V, Litchfield, J.E., Thorpe, S.R. and
D., Sayre, L,M., Monnier, V.M. and Perry G., Advanced Maillard Baynes, J.W., Mechanism of autoxidative glycosylation: identifica-
reaction end products are associated with Alzheimer disase pathol- tion of glyoxal and arabinose as intermediates in the autoxidative
ogy, Proc. Natl. Acad. Sci. USA, 91 Ž1994. 5710–5714. modification of proteins by glucose, Biochemistry, 34 Ž1995. 3702–
w64x Smith, M.A., Sayre, L.M., Monnier. V.M. and Perry, G., Radical 3709.
AGEing in Alzheimer’s disease, Trends Neurosci., 18 Ž1995. 172– w76x Wells-Knecht, M.C., Thorpe, S.R. and Baynes, J.W., Pathways of
176. formation of glycoxidation products during glycation of collagen,
w65x Suarez, G., Etlinger, J.D., Maturana, J. and Weitman, D., Fructated Biochemistry, 34 Ž1995. 15134–15141.
protein is more resistant to ATP-dependent proteolysis than glucated w77x Yan, S.D., Yan, S.F., Chen, X., Fu, J., Chen, M., Kuppusamy, P.,
protein possibly as a result of higher content of Maillard fluoro- Smith, M.A., Perry, G., Godman, G.C., Nawroth, P. and Stern, D.,
phores, Arch. Biochem. Biophys., 321 Ž1995. 209–213. Non-enzymatically glycated t in Alzheimer’s disease induces neu-
w66x Thome, J., Kornhuber, J., Munch,¨ G., Schinzel, R., Taneli, Y., ronal oxidant stress resulting in cytokine gene expression and release
¨
Zielke, B., Rosler, M. and Riederer, P., Eine neue Hypothese zur of amyloid b-peptide, Nature Med., 1 Ž1995. 693–699.
Ätiopathogenese des Alzheimer-Syndroms: advanced glycation end- w78x Yen, S.H., Liu, W.K., Hall, F.L., Yan, S.D., Stern, D. and Dickson,
products ŽAGEs., NerÕenarzt, Ž1996. Žin press.. D.W., Alzheimer neurofibrillary lesions: molecular nature and poten-
w67x Thome, J., Munch,¨ ¨
G., Muller, R., Schinzel, R., Kornhuber, J., tial roles of different components, Neurobiol. Aging, 16 Ž1995.
Blum-Degen, D., Sitzmann, L., Rosler, ¨ M., Heidland, A. and 381–387.
Riederer, P., Advanced glycosylation endproducts-associated param-
eters in the peripheral blood of patients with Alzheimer’s syndrome,
Life Sci., Ž1996. in press.

You might also like