alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced...

23
J Neural Transm (1998) 105: 439–461 Alzheimer’s disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts G. Münch 1 , R. Schinzel 1 , C. Loske 1 , A. Wong 1 , N. Durany 2 , J. J. Li 3 , H. Vlassara 3 , M. A. Smith 4 , G. Perry 4 , and P. Riederer 2 1 Physiological Chemistry, Biocenter, and 2 Clinical Neurochemistry, Department of Psychiatry, University of Würzburg, Federal Republic of Germany 3 The Picower Institute for Medical Research, Manhasset, and 4 Department of Pathology, Case Western Reserve University, Cleveland, OH, USA Accepted March 1, 1998; received November 15, 1997 Summary. Many approaches have been undertaken to understand Alzheimer’s disease (AD) but the heterogeneity of the etiologic factors makes it difficult to define the clincally most important factor determining the onset and progression of the disease. However, there is increasing evidence that the previously so-called “secondary factors” such as a disturbed glucose metabo- lism, oxidative stress and formation of “advanced glycation endproducts” (AGEs) and their interaction in a vicious cycle are also important for the onset and progression of AD. AGEs are protein modifications that contribute to the formation of the histopathological and biochemical hallmarks of AD: amyloid plaques, neurofibrillary tangles and activated microglia. Oxidative modifications are formed by a complex cascade of dehydration, oxidation and cyclisation reactions, subsequent to a non-enzymatic reaction of sugars with amino groups of proteins. Accumulation of AGE-crosslinked proteins throughout life is a general phenomenon of ageing. However, AGEs are more than just markers of ageing since they can also exert adverse biologic effects on tissues and cells, including the activation of intracellular signal transduc- tion pathways, leading to the upregulation of cytokine and free radical pro- duction (oxidative stress). Oxidative stress is involved in various divergent events leading to cell damage, including an increase in membrane rigidity, DNA strand breaks and an impairment in glucose uptake. In addition, other age-related metabolic changes such as depletion of antioxidants or decreased energy production by a disturbed glucose metabolism diminish the ability of the cell to cope with the effects of radical-induced membrane, protein and DNA damage. With our improving understanding of the molecular basis for the clinical symptoms of dementia, it is hoped that the elucidation of the etiologic causes, particularly the positive feedback loops involving radical damage and a reduced glucose metabolism, will help to develop novel

Upload: g-muench

Post on 15-Jul-2016

215 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 439J Neural Transm (1998) 105: 439–461

Alzheimer’s disease – synergistic effects of glucose deficit,oxidative stress and advanced glycation endproducts

G. Münch1, R. Schinzel1, C. Loske1, A. Wong1, N. Durany2, J. J. Li3,H. Vlassara3, M. A. Smith4, G. Perry4, and P. Riederer2

1 Physiological Chemistry, Biocenter, and 2 Clinical Neurochemistry, Department ofPsychiatry, University of Würzburg, Federal Republic of Germany

3 The Picower Institute for Medical Research, Manhasset, and 4 Department ofPathology, Case Western Reserve University, Cleveland, OH, USA

Accepted March 1, 1998; received November 15, 1997

Summary. Many approaches have been undertaken to understandAlzheimer’s disease (AD) but the heterogeneity of the etiologic factors makesit difficult to define the clincally most important factor determining the onsetand progression of the disease. However, there is increasing evidence that thepreviously so-called “secondary factors” such as a disturbed glucose metabo-lism, oxidative stress and formation of “advanced glycation endproducts”(AGEs) and their interaction in a vicious cycle are also important for theonset and progression of AD. AGEs are protein modifications that contributeto the formation of the histopathological and biochemical hallmarks of AD:amyloid plaques, neurofibrillary tangles and activated microglia. Oxidativemodifications are formed by a complex cascade of dehydration, oxidationand cyclisation reactions, subsequent to a non-enzymatic reaction of sugarswith amino groups of proteins. Accumulation of AGE-crosslinked proteinsthroughout life is a general phenomenon of ageing. However, AGEs are morethan just markers of ageing since they can also exert adverse biologic effectson tissues and cells, including the activation of intracellular signal transduc-tion pathways, leading to the upregulation of cytokine and free radical pro-duction (oxidative stress). Oxidative stress is involved in various divergentevents leading to cell damage, including an increase in membrane rigidity,DNA strand breaks and an impairment in glucose uptake. In addition, otherage-related metabolic changes such as depletion of antioxidants or decreasedenergy production by a disturbed glucose metabolism diminish the ability ofthe cell to cope with the effects of radical-induced membrane, protein andDNA damage. With our improving understanding of the molecular basisfor the clinical symptoms of dementia, it is hoped that the elucidation ofthe etiologic causes, particularly the positive feedback loops involvingradical damage and a reduced glucose metabolism, will help to develop novel

Page 2: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

440 G. Münch et al.

“neuroprotective” treatment strategies able to interrupt this vicious cycle ofoxidative stress and energy shortage in AD.

Keywords: Oxidative stress, diabetes, aging, advanced glycation endproducts,lipid peroxidation.

Glucose metabolism in Alzheimer’s disease

One of the striking features of Alzheimer’s disease (AD) is the drastic reduc-tion of glucose metabolism in affected brain areas. In vivo imaging of patientsusing positron emission tomography (PET) with 2-[F-18]-fluoro-2-deoxy-D-glucose as label demonstrates progressive reductions in brain glucose metabo-lism and blood flow in relation to the severity of dementia. Interestingly, notonly patients, but also younger family members with familial AD (FAD;APP717 Val to Ile mutation) show regional brain glucose hypometabolism in apreclinical phase before the onset of the disease, suggesting that changes inthe processing of the amyloid precursor protein (APP) or the ratio of â-amyloid peptide (Aâ) derivatives are linked to the disturbed glucose metabo-lism in AD (Perani et al., 1997). Glucose metabolism in the brain limits thesynthesis of acetylcholine, glutamate, aspartate, gamma-aminobutyric acid,glycine, and ATP production. Whereas the cerebral energy pool is onlyslightly diminished during the normal ageing process, glucose metabolism andcellular energy production are severely reduced in sporadic AD (Hoyer, 1993,1996). This reduced brain glucose utilisation in the early stages of AD reflectspotentially reversible down-regulation of gene expression for oxidative phos-phorylation within neuronal mitochondria (Rapoport et al., 1996). This issupported by the fact that the CSF of AD patients shows higher levels ofpyruvate, which correlates significantly with the severity of dementia (Parnettiet al., 1995).

Although introducing the term “diabetes mellitus of the brain” forAD seems speculative, but nevertheless interesting, there is more andmore evidence for parallel biochemical abnormalities between the glucosehypometabolism in AD and in non-insulin dependent diabetes (NIDDM)(Smith et al., 1996b). High blood and tissue glucose in NIDDM is caused byimpaired insulin signalling (“insulin resistance”) decreasing the ability of theglucose transport system to effectively transport glucose into the cells of theliver and muscle, which leads to a high concentration of extracellular glucoseand AGEs, causing increased oxidative stress (Table 1). Decreased intracellu-lar availability of glucose is particularly detrimental under conditions where itis vital for the cell to increase its cellular antioxidative capacity throughactivation of the pentose phosphate pathway to combat the damaging effectsof oxidative stress. In diabetes, accelerated AGE formation is caused primar-ily by a higher level of plasma glucose. However, accumulation of extracellu-lar AGEs in AD is more likely caused by accelerated oxidation of glycatedproteins (“glycoxidation”), e.g. by redox-active iron bound to proteins inamyloid plaques (Smith et al., 1997). Long-lived protein deposits such asâ-amyloid plaques with bound transition metals create an ideal chemicalenvironment for the formation of AGEs (Wells-Knecht, 1995b). Increased

Page 3: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 441

Table 1. Comparable pathological events in AD and non-insulin dependent diabetesmellitus (NIDDM)

Glycation related factors in NIDDM in AD

Extracellular AGE modified plasma proteins proteins in amyloidproteins (e.g. albumin, plaques (AB, Apo E)

LDL, etc.)

Intracellular AGE modified peripheral neurons: myelin central neurons:proteins basic protein cytoskeletal proteins.

e.g. neurofilamentproteins, microtubuliassociated proteins

Major factor responsible for extracellular: high glucose extracellular: redox-increased AGE formation intracellular: reactive sugars active transition

(fructose-phosphates) metals, e.g. ironintracellular: reactivesugars (fructose-phosphates, trioses)?

Metabolic consequences Oxidative stress, glucose Oxidative stress,hypometabolism and glucoseimpaired cell function hypometabolism and

impaired cell function

accumulation of intracellular AGEs might be caused by an increase in con-centration of more active sugars or sugar fragmentation products (such asfructose and triose phosphates) e.g. by changes in metabolic pathways or ablockade of glycolytic enzymes.

The first type of evidence for the existence of a mechanistic link betweenoxidative stress caused by Aâ and impaired glucose transport was shown incultured neurons by a mechanism involving peroxidation of membrane lipidsleading to a decreased membrane fluidity and loss of protein function. Aâimpairs glucose transport followed by a decrease in cellular ATP levels. Thisimpairment of glucose transport, ATP depletion, and cell death can beprevented by antioxidants. Exposure of cultures to Aâ induces conjugationof 4-hydroxynonenal (HNE), bifunctional carbonyl product of lipid peroxi-dation, to membrane proteins including the GLUT 3 transporter. HNEinduces a concentration-dependent impairment of glucose transport andsubsequent ATP depletion. Lipid peroxidation caused by other sources ofoxidative stress e.g. activated microglia or free extracellular iron may thuscontribute to decreased glucose uptake and neuronal degeneration in AD(Mark et al., 1997). This is consistent with histopathological findings in AD,where a decreased membrane fluidity in mitochondria and an increase of theoxidised nucleoside 8-hydroxy-29-deoxyguanosine in mitochondrial DNA canbe observed, suggestive of a link between oxidative stress and energy produc-tion (Mecocci et al., 1997).

Additional support for parallel biochemical susceptibilities between ADand NIDDM comes from epidemiological data. In two large population based

Page 4: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

442 G. Münch et al.

studies in Rochester and Rotterdam it was convincingly shown, that adult-onset diabetes did not only increase the risk of vascular dementia but also ofAD (Ott et al., 1996; Leibson et al., 1997). Since diminished energy produc-tion by a disturbed glucose metabolism as an age- and disease-associatedchange is very likely to limit cellular capacity to cope with oxidative insults,this might thus be as important from a therapeutic view as the so-called“primary causes” (plaque and tangle formation), which may not be amenableto intervention (Finch and Cohen, 1997).

Advanced glycation endproducts: chemistry

There is more and more evidence that glucose is involved in the pathogenesisof AD in a different way. This involves a non-enzymatic reaction of glucose toform advanced glycation endproducts (AGEs) on long-lived protein deposits,which induce oxidative stress, subsequently disturbing glucose metabolism.AGEs are sugar-derived protein modifications able to irreversibly crosslinklong – lived proteins including the characteristic hallmarks of AD, i.e.â-amyloid plaques and NFT. AGE formation starts with the reaction ofthe amino groups of proteins, particularly the side chains of lysine, arginineand histidine, with reducing sugars, including glucose, fructose, hexose-phosphates, trioses and triose-phosphates. This modification, termed “non-enzymatic glycosylation”, “glycation” or “Maillard reaction”, leads viareversible Schiff-base adducts to protein-bound Amadori products. Throughsubsequent rearrangements, dehydrations and oxidations, a heterogeneousgroup of fluorescent and brown products is formed, the so-called “advancedglycation endproducts” (AGEs) (Brownlee, 1995; Buccala and Cerami, 1992).AGEs can be formed by non-oxidative and oxidative reaction pathways, thelatter being significantly accelerated by transition metals, such as copper andiron. In this “glycoxidation” reaction, protein-bound AGEs as well as soluble,highly reactive dicarbonyl products and oxygen free radicals are formed(Wells-Knecht et al., 1995a). Transition metals can also oxidise the monosac-charides directly in solution to form dicarbonyl products, which subsequentlycrosslink proteins through a process called “autooxidative glycosylation”. Theoxidation steps in both pathways can be inhibited by metal chelators,emphasising the significance of transition metals for AGE-formation (Wells-Knecht et al., 1995b). Among the physiologically relevant sugars, glucose isthe least reactive, presumably the reason for its selection by evolution as themain biological energy carrier. The rank order of reactivity for the othermonosaccharides increases from hexoses to trioses by several orders of mag-nitude (Namiki, 1988). AGE formation is irreversible and causes protease-resistant cross-linking of peptides and proteins, leading to protein depositionand amyloidosis (Smith et al., 1994b, 1995; Vitek et al., 1994) (Fig. 1).

Interaction of AGEs with cells

It is becoming increasingly clear how AGEs are involved in the formation ofthe abnormal filamentous lesions, NFT and senile plaques, in and outsideneurons in the brains of afflicted individuals. However, the effects of AGEs

Page 5: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 443

Fig. 1. Chemical reactions leading to the formation of advanced glycation endproducts

on cells, particularly their interaction with cell surface receptors and intracel-lular signal transduction, is a relatively new direction of research which mighthelp to solve certain aspects of the etiopathogenesis of AD. While it has beenargued a few years ago that the AGEs on these deposits are static by-productsof the disease rather than dynamic participants in neuronal death, it is becom-ing more and more obvious that AGEs in senile plaques and NFT exertmultiple detrimental effects on cells. For example:

Glycated proteins produce radicals through oxidation reactions

Formation of oxygen free radicals is associated with the Maillard reaction.Glycated proteins produce nearly 50 fold more radicals than non-glycatedproteins (Mullarkey et al., 1990). This process commences with the produc-tion of superoxide radicals by the transition metal-catalysed autoxidation ofprotein-bound Amadori products, followed by the dismutation of superoxideto hydrogen peroxide, and the generation of hydroxyl radicals by the Fentonreaction. This results in a site-specific attack on proteins, with consequentprotein damage and lipid peroxidation, as well as damage to other cell com-ponents, such as DNA (Smith et al., 1995).

AGEs activate microglia and induce cytokine production andrelease of oxygen free radicals

Interaction of AGEs with cells increases oxidative stress. It is not yet clearwhether this occurs simply by the binding of AGEs to the cell surface andsubsequent diffusion of chemically produced free radicals across the mem-

Page 6: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

444 G. Münch et al.

brane, or by a receptor-mediated oxidative signalling pathway. In addition tothe recently described AGE-receptor, RAGE (Schmidt et al., 1993, 1994),various other receptors with AGE binding properties have been described(Vlassara et al., 1995). Interestingly, the macrophage scavenger receptorwhich is associated with senile plaques in AD (Christie et al., 1996), alsomediates the endocytic uptake and degradation of AGE proteins (Araki et al.,1995). These AGE-mediated oxidative signalling effects can be blocked bythe addition of antioxidants and antibodies to the AGE-receptor (Wautier etal., 1994). In cell culture experiments, PHF-tau isolated from post-mortemtissue and AGE- modified recombinant MAP-tau each generate oxygen freeradicals, thereby not only activating transcription via NFκB, but also inducingthe release of the characteristic 4kDa Aâ peptides (Schmidt et al., 1994;Yan et al., 1995).

What may be the most important contribution of AGEs to theetiopathogenesis of AD is free radical production via the innate immunesystem. It has been shown by several groups that AGE-modified proteins canelicit an acute phase response in microglial cells, generate a “respiratoryburst” and cause “bystander-lysis” of neighbouring neurons (McMillian et al.,1995). These findings corroborate with earlier studies showing that AGEsinduce chemotaxis (Schmidt et al., 1993) and inflammatory responses in mac-rophages including secretion of interleukin 1 and 6, and TNF-α (Vlassara etal., 1988; Guzdek and Stalinska, 1992) (Fig. 2).

The Aâ deposition in Alzheimer’s disease bears similarity to otheramyloidoses such as the hemodialysis-associated amyloidosis (HAA), a com-plication of long-term hemodialysis whereby â2-microglobulin (â2M) is depos-ited in â-sheeted amyloid fibrils. â2M in the amyloid deposits of HAA is alsomodified by AGEs. AGE-â2M enhances directed migration (chemotaxis) andrandom cell migration (chemokinesis) of human monocytes. In addition,AGE-â2M increases the secretion of TNF-α and IL-1â from macrophages.These findings suggest that AGE-â2M, a major component of amyloiddeposits, participates in the pathogenesis of HAA as foci where monocytes/macrophages accumulation initiates an inflammatory response that leads tobone/joint destruction (Miyata et al., 1996).

If a pathophysiological cascade similar to HAA is also occurring in AD, itmight explain why such an autodestructive process, involving reactive astro-cytes and microglial cells, reflected by an increase in pro-inflammatorycytokines in the CSF (Blum-Degen et al., 1995), exists in the AD brain(McGeer et al., 1994).

AGE receptors – physiological role and pathophysiological effects

AGEs are markers of protein ageing. In view of the close association of AGEswith cells in the body and their known adverse biologic effects on cells, manystudies have focused on searching for AGE binding proteins. The rationale isthat AGE binding proteins might play important roles in the removal ofAGEs from the tissue, thus indirectly determining the ageing process per se inthe body (Monnier and Cerami, 1981; Monnier et al., 1991).

Page 7: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 445

Fig. 2. Direct and indirect toxic effects of AGEs through crosslinking of Aâ peptide

Binding and internalisation of AGE-modified proteins is mediatedby several AGE- specific cell surface receptors, including AGE-R1 (OST-48/p60), AGE-R2 (80K-H/p90) and AGE-R3 (galectin-3) (Yang et al., 1991;Vlassara et al., 1995; Li et al., 1996), scavenger-receptor (Smedsrod et al.,1997), and a receptor for AGEs (RAGE) (Schmidt et al., 1996; Miyata et al.,1996; Yan et al., 1996). These receptors are widely distributed amongdifferent cell types including hemopoetic, endothelial, mesangial, neuronaland glial cells (Imani et al., 1993; Li et al., 1996; Yan et al., 1996). There isincreasing evidence that AGE receptors play important roles in the catabo-lism of AGEs in the body (Yang et al., 1991). Smedsrod et al. show that ratliver scavenger receptors are capable of eliminating AGE-modified bovineserum albumin from the blood (Smedsrod et al., 1997). We also have evidencesuggesting that AGE receptors on astrocytes participate in the degradation ofAGE-modified proteins in human brain (Li et al., 1998). It has also beenshown that AGE-R1-3 expression is a dynamic process and can be up-

Page 8: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

446 G. Münch et al.

regulated by cytokines (Vlassara et al., 1989). AGE-R3 expression can beenhanced by GM-CSF in human mononuclear phagocytes, but not in astro-cytes (Li et al., 1998). Our results corroborate well with the fact that GM-CSFcan up – regulate AGE-R3 (MAC-2) expression in macrophages and Schwanncells (Saada et al., 1996). The most interesting AGE receptor due to its abilityto take part in signal transduction across the membrane is RAGE (Yan et al.,1996; Bierhaus et al., 1997). In addition to binding to AGE-modified proteins,RAGE can also interact with amphoterin to regulate neurite outgrowthduring nerve development (Hori et al., 1995), and with â-amyloid peptide toinduce intracellular oxidative stress and to activate NFκB regulated geneexpression (Yan et al., 1996).

Cellular signalling of AGEs has recently emerged as an important linkin the pathophysiological events of AD, showing activation of cellular tran-scription mechanisms in the response to an elevated AGE microenviroment.AGEs have been shown to activate two major signal transduction pathways:a redox-sensitive pathway involving the transcription factor NFκB (Schmidtet al., 1996) and a mitogenic pathway involving protein kinases includingERK-2 and the transcription factor AP-1 (Lander et al., 1997; Simm et al.,1997) (Fig. 3). Both signal transduction pathways could be involved in thepathophysiological events in AD.

NADPH-oxidase/superoxide/NFκB-pathway

The redox-sensitive NFκB pathway is likely to be important in neurons,astroglia and microglia where AGEs or Aâ might cause the release ofcytokines and free radicals. Activation of the p65 NFκB subunit was observedin the neurons and astroglia of brain sections from patients with AD, wherebyactivated NFκB was shown to be restricted to cells in the close vicinity of earlysenile plaques (Kaltschmidt et al., 1997). The NADPH-oxidase/superoxide/NFκB-pathway can be initiated by both AGEs and Aâ and uses reactiveoxygen intermediates as messengers (Yan et al., 1996). Maximal activation ofNFκB requires 0.1µM Aâ (1–40) which is about 10–20 times less than the EC50

for direct neurotoxicity (Kaltschmidt et al., 1997). A similar ratio betweenactivation and toxicity thresholds can be observed with model-AGEs whereupregulation of the inducible nitric oxide synthase, a NFκB regulated gene, isabout 50 times lower than the AGE concentration necessary for the inductionof significant cell death (Münch et al., 1997b).

p21(ras)/MAP-kinase (ERK) pathway

Studies in rat pulmonary artery smooth muscle cells have shown that AGEsare able to activate p21(ras) as well as the MAP-kinases Erk 1 and ERK2 viaa RAGE-dependent pathway (Lander et al., 1997). This result is supported bystudies in the renal tubulus cell line LLC-PK1, where AGEs were shown toactivate p42(MAP) kinase (ERK 2) and its downstream target, the AP-1complex (Simm et al., 1997). This might explain one of the paradox findings inAD, namely the activation of mitogenic signal transduction pathways in ADneurons. For example, the expression of the small G-protein p21(ras), a

Page 9: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 447

critical regulator of cell proliferation and differentiation is increased withinneuritic plaques as well as in neurons and glial cells closely associated withplaques. Neurons containing tangle-bearing material also show a high level ofp21(ras) expression (Gärtner et al., 1995). The downstream members of thep21(ras) pathway are the mitogen-activated protein kinase kinase (MAPK-kinase) and its substrate, the mitogen-activated protein kinase (MAP-kinaseor ERK), which is able to phosphorylate MAP-tau, thereby generating abnor-mally hyperphosphorylated tau species that are similar to PHF-tau found inAD. Immunoreactive neurons are localized in the direct vicinity of neuriticsenile plaques (Arendt et al., 1995). In addition, the cyclin-dependent kinaseinhibitor p16, a regulator of the orderly progression through the cell cycle, isaccumulated in both neurofibrillary tangles and neuritic components of senileplaques (Arendt et al., 1996; McShea et al., 1997). A second important regu-lator of the cell cycle, cyclin-dependent kinase-4 (cdk-4) was shown to beincreased in the brains of cases of AD patients compared with age-matchedcontrols. Both proteins are increased in the pyramidal neurons of the hippo-

Fig. 3. Schematic illustration of putative AGE-induced signal transduction pathways

Page 10: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

448 G. Münch et al.

campus, including those neurons containing NFT and granulovacuolar degen-eration (McShea et al., 1997). As p16 is not normally found in terminallydifferentiated neurons, it seems paradoxical that it is increased in AD. Induc-tion of p16, a protein that signals re-entry and progression through the cellcycle, may itself be the consequence of a response to a growth stimulus. Re-entry into the cell cycle is likely deleterious in terminally differentiated neu-rons and may contribute to the biochemical abnormalities, particularly axonalsprouting and hyperphosphorylation of tau as well as the neuronal degenera-tion characteristic of the pathology of AD (McShea et al., 1998). Thus it isvery likely that the activation of the MAP kinase cascade by AGEs is criticallyinvolved in the processes of neurodegeneration and aberrant repair.

Advanced glycation endproducts in AD

AGEs accumulate in pyramidal (glutaminergic) neurons in aged animals andhumans, but their intracellular distribution pattern is quite different betweenanimal and man. In the human pyramidal neuron, AGEs exhibit a granular,perikaryonal distribution, whereas in animal brains, AGEs show a nuclearstaining pattern (Li et al., 1994). Pyramidal neurons selectively accumulateAGE-containing vesicles in an age-dependent manner presumably in endo-somes or lysosomes (Kimura et al., 1996; Li et al., 1995). In addition to intra-cellular cytoplasmic accumulation of AGEs during ageing, proteins presentin senile plaques and NFT have been shown to contain significant amountsof AGEs:

Senile plaques

The major proteinaceous component of the amyloid deposits which accumu-late extracellularly in the AD brain, is Aâ. This 39–42 amino acid peptideconsists of 28 extramembrananal amino acids plus 11 to 14 residues of thehydrophobic transmembrane domain of its precursor, amyloid precursor pro-tein, APP (Price and Sisodia, 1994). Genetic studies have linked some familialcases of early onset Alzheimer’s disease to the APP-locus on chromosome 21(,1% of all AD cases), where mutations in the APP-gene influence theprocessing of APP with subsequent changes in Aâ concentration or the ratioof the 1–42 to 1–40 Aâ form (Hardy, 1997; Citron et al., 1997). In analogy,mutations in the presenilin genes change the ratio of the 1–42 to 1–40 Aâform, most likely by influencing APP processing in the endoplasmatic reticu-lum (Hartmann et al., 1997).

Two major biochemical mechanisms have been proposed by which Aâmay be involved in neuronal cell death: direct neurotoxicity and/or indirect,immune system-mediated neurotoxicity as the result of microglial cell activa-tion. Firstly, several studies have demonstrated that Aâ peptide is both toxicto neurons in primary culture and to neuronal cell lines (Lorenzo andYankner, 1996; Zhang et al., 1996). The neurotoxic effect appears to correlatewith the aggregation state and degree of â-sheet conformation (Pike et al.,1995). In addition, oxidative stress (Behl et al., 1994), apoptosis (Loo et al.,1993) and ion channel formation (Pollard et al., 1995) can combine to mediate

Page 11: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 449

Aâ neurotoxicity. However, it appears that overexpression of APP intransgenic mice with resulting Aâ deposition does not cause neuronal celldeath and behavioural changes related to “dementia”, although the â-amyloiddeposits are associated with gliosis and neuritic dystrophy, suggesting thatadditional factors are necessary to promote the progression of the disease(Irizarry et al., 1997). One of these additional factors may be the activation ofresting microglia cells by the Aâ deposits; as a result, many inflammatorymediators, such as oxygen free radicals, NO, cytokines and complementproteins are released. These inflammatory mediators can subsequently dam-age surrounding neurons through “bystander lysis” (McGeer et al., 1994,1997).

AGE accumulation has been demonstrated in senile plaques in differentcortical areas (Smith et al., 1994b,c; Vitek et al., 1994) in primitive plaques,coronas of classic plaques and some glial cells of AD brain (Kimura et al.,1995). The higher AGE level in AD tissue is reflected by increased AGE-Aâlevels in the CSF of AD patients (Li J. et al., unpublished data). We and othershave demonstrated in vitro that nucleation-dependent polymerisation of Aâ,the major component of senile plaques is significantly accelerated by AGE-mediated crosslinking (Vitek et al., 1994; Münch et al., 1997a). Aâ aggrega-tion follows a classic nucleation-dependent polymerisation process, consistingof two distinctive steps, an initial slow nucleus formation step, followed by arapid elongation phase (Jarett and Lansbury, 1993). The slow nucleus forma-tion stage is a reversible process depending on the concentration of themonomer. Therefore, at a sub-threshold peptide concentration, irreversiblecovalent crosslinking by AGEs play a crucial role in stabilising monomernucleation as well as growth of the larger aggregates, the amyloidplaques. Since the aggregation of Aâ therefore is critical for its toxiceffects via the stimulation of either direct or indirect neurotoxicity, the rate ofsenile plaque formation is likely to be important for the progression ofthe disease. This suggests that AGEs may indeed represent a driving force inthe acceleration of â-amyloid deposition and senile plaque formation (Smithet al., 1995).

It is now well accepted that AD susceptibility is correlated with the doseof the ε4 allele of apolipoprotein E (Apo E) (Czech et al., 1995). Substitutionof cysteines by arginines creates more glycation sites and hence, facilitates theAGE-mediated crosslinking of ApoE to the insoluble amyloid fibrils andaccelerates senile plaque formation (Harrington and Colaco, 1994; Li andDickson, 1997).

Neurofibrillary tangles (NFTs)

NFTs and neuropil threads are also histopathological hallmarks of AD. Theinterrelationships between the rate of NFT formation and the progression ofAD remain unknown (Cras et al., 1995). Nonetheless, early signs of tangleformation in certain brain regions such as the entorhinal cortex precede theclinical diagnosis of AD by decades (Braak and Braak, 1995). The majorcomponent of NFT is hyperphosphorylated microtubule-associated protein

Page 12: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

450 G. Münch et al.

tau (MAP-tau). This abnormal MAP-tau is resistant to proteolytic enzymesand it is suggested that glycation, disulphide bond formation, phosphorylation(Smith et al., 1994c, 1995; Mandelkow et al., 1996; Schweers et al., 1995) and/or formation of core fragments of MAP-tau (Novak et al., 1993) can allcontribute to extensive cross-linking between MAP-tau subunits.

NFT isolated from brain tissue of AD patients are detergent-insoluble andprotease-resistant and have characteristic fluorescent spectra of brown pig-ments similar to synthetic AGEs (Colaco et al., 1996). The major componentof the NFT is MAP-tau, which has been shown to be subject to intracellularglycation and AGE formation (Ledesma et al., 1994). The relevance ofglycation as an additional pathological modification of MAP-tau in vitro issupported by the immunohistochemical co-localization of AGEs with NFT(Smith et al., 1994c; Dickson et al., 1996). MAP-tau can be glycated in vitro,inhibiting its ability to bind to microtubules; in addition, paired helical fila-ment (PHT)-tau isolated from the brains of AD patients is glycated in thetubulin-binding region (Ledesma et al., 1995). Interestingly, only the glycationof MAP-tau gives rise to the formation of fibrils which resemble NFT in theAD brain (Colaco et al., 1996). Finally, the introduction of glycated MAP-tau into cells can generate oxygen-free radicals capable of disturbingneuronal function, including up-regulation of APP and release of Aâ (Yanet al., 1995).

Oxidative stress in Alzheimer’s disease

The first hints for oxidative stress in AD came from studies of fibroblasts fromAD patients where several key enzyme abnormalities were found. Two oxida-tive abnormalities were consistently noted; deficiencies in thiamine-requiringenzymes (Gibson et al., 1988) and changes in the mitochondrial respiratorychain (Sheu et al., 1994). Uncoupling of electron transport from oxidativephosphorylation leads to incomplete reduction of oxygen, producing straysuperoxide radicals. The addition of chemical uncoupling agents to neuroblas-toma cell lines leads to the appearance of abnormal filaments similar to thoseforming NFT (Blass et al., 1990). These findings have been applied to rodentsby Gibson and co-workers who fed rats or mice a thiamine-deficient diet andfound clusters of dystrophic neurites similar to those in senile plaques in AD(Calingasan et al., 1995, 1996). That oxidatively-compromised animals de-velop AD-type neuritic dystrophy suggests that disturbed energy metabolismand subsequent oxidative stress may be a common denominator of neuriticdystrophy. Recent studies suggest that oxidative stress may also play a role inDown’s syndrome which is trisomic for superoxide dismutase. Moreover,neurons in culture from Down’s syndrome cases undergo a prematureapoptotic death that is delayed by antioxidants (Busciglio and Yankner,1995).

There is overwhelming evidence that tissue in AD is exposed to oxidativestress during the course of the disease (Fig. 4). NFT bear the footprints ofoxidative membrane damage since they contain adducts of malondialdehyde,the most well known lipid peroxidation product (Yan et al., 1994), and

Page 13: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 451

Fig. 4. Sources and effects of oxidative stress on a molecular and cellular level

hydroxynonenal, the most highly reactive lipid peroxidation product(Montine et al., 1996a; Sayre et al., 1997). Furthermore, dystrophic neurites ofsenile plaques that contain the PHF filaments also show greater membranedamage than those that lack abnormal filaments (Praprotnik et al., 1996).

AD tissue shows an imbalance in radical detoxifying enzymes. The ratio ofsuperoxide dismutase to catalase decreases, leading to a buildup of hydrogenperoxide, which is particularly dangerous in the presence of free iron due tosubsequent production of hydroxyl radicals (Götz et al., 1994; Gerlach et al.,1994; Gsell et al., 1995).

Increased levels of DNA strand breaks have been found in AD using theTUNEL labelling technique.While DNA strand cleavage by endonucleases isa part of apoptosis, strand breaks in AD are found in nearly all brain cells,which is surprising for a chronic condition (Anderson et al., 1996). Therefore,it is likely that oxidative damage to DNA, rather than apoptosis, is responsiblefor DNA breakage in AD and this is consistent with the increased freecarbonyls in the nuclei of neurons and glia in AD (Smith et al., 1996a).

The induction of heme oxygenase-1, an antioxidant enzyme involved inthe conversion of heme to bilirubin, is a robust indicator of the oxidative stressresponse in cells (Maines, 1988). In previous studies, we demonstratedthat the heme oxygenase-1 protein (Smith et al., 1994a, 1995) and mRNA(Premkumar et al., 1995) were increased in AD brains and this increase wastightly correlated with regions of NFT pathology. Furthermore, glycated taufound in neurons in AD (Smith et al., 1994b; Yan et al., 1994; Ledesma et al.,1994) causes an oxidative response in neuroblastoma cells including inductionof heme oxygenase-1, translocation of NFκB and peroxidation of membranelipids (Yan et al., 1994, 1995).

Evidence continues to mount that bifunctional 4-hydroxy-2-alkenals, suchas 4-hydroxy-2-nonenal (HNE), as opposed to other reactive lipid-derivedaldehydes such as malondialdehyde (MDA) or 2-alkenals, are the majorcytotoxic products of lipid peroxidation (Esterbauer et al., 1991) (Fig. 5).Following lipid peroxidation, a 2-pentylpyrrole modification of lysine is theonly presently known “advanced” (stable end-product) adduct that formsfrom the modification of proteins by HNE (Sayre et al., 1993). Rabbit

Page 14: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

452 G. Münch et al.

Fig. 5. Reactions scheme for the formation of HNE modified proteins

polyclonal antibodies specific to this HNE-pyrrole modification label not onlyneurofibrillary pathology but also pyramidal neurons in cases of AD, but notcontrol patients (Sayre et al., 1997). These findings, together with the recentdemonstration that HNE is cytotoxic to neurons (Montine et al., 1996a,b) andthat it impairs the function of membrane proteins including the neuronalglucose transporter 3 (GLUT 3) (Mark et al., 1997), indicate that HNEis not only a characteristic marker but also a major toxin leading toneurodegenerative events in AD.

Since oxidative stress is characterized by the imbalance of radical produc-tion and antioxidative defense, the latter is considered to be also a majorplayer in the process of age-related neurodegeneration. Reducing equivalentsare responsible for the endogenous redox potential in the living cell which iskept under tight regulation. Among their various roles is the ability to donateelectrons to reactive oxygen species and by doing so, to scavenge them. Mostof the reducing equivalents in the cell are molecules with low molecularweights, such as glutathione and NADH which are synthesized by the cell(Kohen et al., 1997).

It has been shown that the antioxidant cell defense system increaseswith age but the rate of reactive oxygen species generation exceeds theinduced antioxidant ability, generating a situation that favours oxidativestress and peroxidation. The decrease in the concentration of reduced glu-tathione is dramatic; it reaches up to 50% (human erythrocytes) or 70% (rathepathocytes) (Richards et al., 1997; Sanz et al., 1997).

Pharmacological interference with AGE formation or signalling as anovel treatment strategy for AD

The focus of drug development for AD remains at a very unsatisfying state,primarily to an emphasis on pallative treatments with cholinesterase-inhibitors or cholinergic agonists. However, pharmacological approachestargeting oxidative stress, protein glycation, glucose metabolism and the acute

Page 15: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 453

phase response offer new opportunities for the treatment of AD (Heidrichet al., 1997).

AGE-Inhibitors

Inhibitors of the Maillard reaction such as aminoguanidine are currently beingassessed in clinical trials for the treatment of diabetic complications. In con-trast, their potential for the treatment of AD is only just being recognised. TheAGE-inhibitor Tenilsetam has been shown to improve cognitive abilities andmemory constantly over a time span of three months in two phase II trials(Dierks et al., 1989; Ihl et al., 1989; Münch et al., 1994; Shoda et al., 1997).These recent studies indicate possible neuroprotective approaches to thedisease, based on inhibition of the action of AGEs by the use of AGE-bindingcompounds and the modulation of degradation of AGE-cross-linked proteins(Colaco and Harrington, 1996; Münch et al., 1997b).

Anti-inflammatory drugs

Similar beneficial results for AD patients are seen with the non-steroidal anti-inflammatory drugs (NSAIDs) indomethacin, ibuprofen, tenidap and acetyl-salicylic acid (Lieb et al., 1997). Although a direct inhibitory role of theNSAIDs on glycation is discussed (Colaco and Harrington, 1996; Colaco et al.,1996), their primary targets are signal transduction pathway involvingprostaglandins, e.g. between AGE-receptors and downstream effectors in-volved in cytokine and radical production (Gonzales et al., 1994; McGeer andMcGeer, 1997).

Antioxidants

There is evidence that vitamins that protect against oxidative damage orinhibit radical producing enzymes may reduce the neuronal damage and slowthe progression of AD (Frölich and Riederer, 1995). In a recent controversialstudy, patients with moderate AD receiving the monoamine oxidase B(MAO-B) inhibitor selegiline, α-tocopherol (vitamin E, 2000IU per day)were shown to deteriorate slower than the respective placebo group (Sano etal., 1997). In addition, two novel antioxidants possess not merely passiveradical scavenging capabilities, but can also block the redox-sensitive NFκBmediated signal transduction pathways of AGEs. One of them, thioctic (α-lipoic) acid, combines antioxidant with energy enhancing effects, includingupregulation of glucose metabolism, direct radical scavenging, redox modula-tion of cell metabolism, and the potential to inhibit oxidatively-induced injury.Reduction of lipoate to dihydrolipoate occurs by specific NADH andNADPH dependent enzymes and is an important requirement (Haramaki etal., 1997; Roy et al., 1997), since only the reduced form of thioctic acid inhibitsAGE-induced activation of NFκB (Bierhaus et al., 1997). A second class ofnovel and promising antioxidants are estrogen derivatives such as 17â-estradiol, which are excellent candidates for intracellular radical scavenging(Behl et al., 1997). This might also explain the reduced risk for AD in post-

Page 16: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

454 G. Münch et al.

menopausal women undergoing estrogen replacement therapy (Ohkura et al.,1994).

Although it remains a very speculative hypothesis, interference withAGE-induced signalling might explain the success of some of the treatmentstrategies mentioned above. AGE inhibitors might be able to stop formationof AGE-Aâ deposits or modify their structure causing loss of binding to AGEreceptors, thus interrupting the AGE-induced signal transduction pathway atthe earliest possible step. Antioxidants scavenge intracellular and extracellu-lar radicals before they damage cell constituents or, especially in microglia,induce cytokine and radical production (Yan et al., 1996). A similar interfer-ence with proinflammatory signal transduction might be the predominant wayof action for the NSIADs. However, future studies need to prove that thesesubstances are truely neuroprotective, demonstrated not only by improve-ment of cognitive function in AD patients, but also by a decelerated rate ofneural cell death, evident by low levels of MAP-tau (released from dyingcells) in the CSF or by a delayed progression of lesion formation monitored byimaging techniques.

Acknowledgements

We thank A. Hipkiss, J. Michaelis, R. Holliday, U. Schindler, A. M. Cunningham, G.Grigg, F. Pemper and D. Palm for valuable and critical discussions and the “Claussen-Stiftung” and the “Hirnliga e.V.” for funding. This work was supported by a grant fromBIOMED I Nr. BMH 1-CT 94-1563.

References

Anderson AJ, Su JH, Cotman CW (1996) DNA damage and apoptosis in Alzheimersdisease – colocalization with c-jun immunoreactivity, relationship to brain area, andeffect of postmortem delay. J Neurosci 16: 1710–1719

Araki N, Higashi T, Mori T, Shibayama R, Kawabe Y, Kodama T, Takahashi K, ShichiriM, Horiuchi S (1995) Macrophage scavenge receptor mediates the endocytic uptakeand degradation of advanced glycation end products of the Maillard reaction. Eur JBiochem 230: 408–415

Arendt T, Holzer M, Grossmann A, Zedlick D, Bruckner MK (1995) Increased expres-sion and subcellular translocation of the mitogen activated protein kinase kinase andmitogen-activated protein kinase in Alzheimers disease. Neuroscience 68: 5–18

Arendt T, Rödel L, Gärtner U, Holzer M (1996) Expression of the cyclin-dependentkinase inhibitor p16 in Alzheimers disease. Neuroreport 7: 3047–3049

Behl C, Davis JB, Lesley R, Schubert D (1994) Hydrogen peroxide mediates amyloid betaprotein toxicity. Cell 77: 817–827

Behl C, Skutella T, Lezoualch F, Post A, Widmann M, Newton CJ, Holsboer F (1997)Neuroprotection against oxidative stress by estrogens – structure-activity relation-ship. Mol Pharmacol 51: 535–541

Bierhaus A, Illmer T, Kasper M, Luther T, Quehenberger P, Tritschler H, Wahl P,Ziegler R, Müller M, Nawroth PP (1997) Advanced glycation end product (AGE)-mediated induction of tissue factor in cultured endothelial cells is dependent onRAGE. Circulation 96: 2262–2271

Blass JP, Baker AC, Ko L, Black RS (1990) Induction of Alzheimer antigens by anuncoupler of oxidative phosphorylation. Arch Neurol 47: 864–869

Blum-Degen D, Müller T, Kuhn W, Gerlach M, Przuntek H, Riederer P(1995) Interleukin-1-beta and interleukin-6 are elevated in the cerebrospinal

Page 17: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 455

fluid of Alzheimers and de novo parkinsons disease patients. Neurosci Lett 202: 17–20

Braak H, Braak E (1995) Staging of Alzheimer’s disease-related neurofibrillary changes.Neurobiol Aging 16: 271–278

Brownlee M (1995) Advanced protein glycosylation in diabetes and aging. Annu RevMed 46: 223–234

Buccala R, Cerami A (1992) Advanced glycosylation: chemistry, biology, and implica-tions for diabetes and aging. Adv Pharmacol 23: 1–34

Busciglio J, Yankner BA (1995) Apoptosis and increased generation of reactive oxygenspecies in downs syndrome neurons in vitro. Nature 378: 776–779

Calingasan NY, Gandy SE, Baker H, Sheu KFR, Kim KS, Wisniewski HM, Gibson GE(1995) Accumulation of amyloid precursor protein-like immunoreactivity in rat brainin response to thiamine deficiency. Brain Res 677: 50–60

Calingasan NY, Gandy SE, Baker H, Sheu KFR, Smith JD, Lamb BT, Gearhart JD,Buxbaum JD, Harper C, Selkoe DJ, Price DL, Sisodia SS, Gibson GE (1996) Novelneuritic clusters with accumulations of amyloid precursor protein and amyloidprecursor-like protein 2 immunoreactivity in brain regions damaged by thiaminedeficiency. Am J Path 149: 1063–1071

Christie RH, Freeman M, Hyman BT (1996) Expression of the macrophage scavengerreceptor, a multifunctional lipoprotein receptor, in microglia associated with senileplaques in Alzheimer’s disease. Am J Pathol 14: 8399–8403

Citron M, Westaway D, Xia WM, Carlson G, Diehl T, Levesque G, Johnsonwood K, LeeM, Seubert P, Davis A, Kholodenko D, Motter R, Sherrington R, Perry B, Yao H,Strome R, Lieberburg I, Rommens J, Kim S, Schenk D, Fraser P, Hyslop PS, SelkoeDJ (1997) Mutant presenilins of Alzheimers disease increase production of 42-residue amyloid beta-protein in both transfected cells and transgenic mice. NatureMed 3: 67–72

Colaco CALS, Harrington CR (1996) Inhibitors of the Maillard reaction – potential in thetreatment of Alzheimers disease. CNS Drugs 6: 167–177

Colaco CALS, Ledesma MD, Harrington CR, Avila J (1996) The role of the Maillardreaction in other pathologies – Alzheimers disease. Nephrol Dial Transplant 11: 7–12

Cras P, Smith MA, Richey PL, Siedlak SL, Mulvihill P, Perry G (1995) Extracellularneurofibrillary tangles reflect neuronal loss and provide further evidence of extensiveprotein cross-linking in Alzheimer disease. Acta Neuropathol 89: 291–295

Czech C, Förstl H, Hentschel F, Monning U, Besthorn C, Geiger-Kabisch C, Sattel H,Masters C, Beyreuther K (1995) Apolipoprotein ε-4 gene dose in clinically diagnosedAlzheimer’s disease: prevalence, plasma cholesterol levels and cerebrovascularchange. Eur Arch Psychiat Clin Neurosci 243: 291–292

Dickson DW, Sinicropi S, Yen SH, Ko LW, Mattiace LA, Bucala R, Vlassara H (1996)Glycation and microglial reaction in lesions of Alzheimer’s disease. Neurobiol Aging17: 733–743

Dierks T, Maurer K, Ihl R (1989) Influence of tenilsetam on AEP-300 in Alzheimer’sdisease. J Neural Transm [P-D-Sect] 1: 49

Esterbauer H, Schaur RJ, Zollner H (1991) Chemistry and biochemistry of 4-hydroxynonenal, malonaldehyde and related aldehydes. Free Radic Biol Med 11: 81–128

Finch CE, Cohen DM (1997) Aging, metabolism, and Alzheimer disease – review andhypotheses. Exp Neurol 143: 82–102

Frölich L, Riederer P (1995) Free radical mechanisms in dementia of Alzheimer type andthe potential for antioxidative treatment. Drug Res 45: 443–336

Gärtner U, Holzer M, Heumann R, Arendt T (1995) Induction of p21ras in Alzheimerpathology. Neuroreport 6: 1441–1444

Gerlach M, Ben-Shachar D, Riederer P, Youdim MBH (1994) Altered brain metabolismof iron as a cause of neurodegenerative diseases? J Neurochem 63: 793–807

Page 18: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

456 G. Münch et al.

Gibson GE, Sheu KF, Blass JP, Baker A, Carlson KC, Harding B, Perrino P (1988)Reduced activities of thiamine-dependent enzymes in the brains and peripheraltissues of patients with Alzheimer’s disease. Arch Neurol 45: 836–840

Götz ME, Künig G, Riederer P, Youdim MBH (1994) Oxidative stress: free radicalproduction in neural degeneration. Pharm Ther 63: 37–122

Gonzalez E, de la Cruz C, de Nicolas R, Egido J, Herrero-Beaumont G (1994) Long-termeffect of nonsteroidal anti-inflammatorydrugs on the production of cytokines andotherinflammatory mediators by blood cells of patients withosteoarthritis. AgentsActions 41: 171–178

Gsell W, Conrad R, Hickethier M, Sofic E, Frölich L, Wichart I, Jellinger K, Moll G,Ransmayr G, Beckmann H, Riederer P (1995) Decreased catalase activity butunchanged superoxide dismutase activity in brains of patients with dementia ofAlzheimer type. J Neurochem 64: 1216–1223

Guzdek A, Stalinska K (1992) Glycated proteins as inducers of acute phase cytokines.Fol Histochem Cytobiol 30: 167–169

Haramaki N, Han D, Handelman GJ, Tritschler HJ, Packer L (1997) Cytosolic andmitochondrial systems for NADH- and NADPH-dependent reduction of alpha-lipoicacid. Free Radic Biol Med 22: 535–542

Hardy J (1997) Amyloid, the presenilins and Alzheimers disease. Trend Neurosci 20: 154–159

Harrington CR, Colaco CA (1994) Alzheimer’s disease. A glycation connection. Nature370: 247–248

Hartmann T, Bieger SC, Bruhl B, Tienari PJ, Ida N, Allsop D, Roberts GW, Masters CL,Dotti CG, Unsicker K, Beyreuther K (1997) Distinct sites of intracellular productionfor Alzheimers disease Aâ-40/42 amyloid peptides. Nature Med 3: 1016–1020

Heidrich A, Rösler M, Riederer P (1997) Pharmacotherapy in Alzheimers dementia –treatment of cognitive symptoms – results of new studies. Fortschr Neurol Psychiat65: 108–121

Hori O, Brett J, Slattery T, Cao R, Zhang J, Chen JX, Nagashima N, Lundh ER, Vijay S,Nitecki D (1995) The receptor for advanced glycation end products (RAGE) is acellular binding site for amphoterin. Mediation of neuritic outgrowth and co-expression of RAGE and amphoterin in the developing nervous system. J Biol Chem270: 25752–25761

Hoyer S (1993) Abnormalities in brain glucose utilization and its impact on cellular andmolecular mechanisms in sporadic dementia of Alzheimer type. Ann NY Acad Sci695: 77–80

Hoyer S (1996) Oxidative metabolism deficiencies in brains of patients with Alzheimersdisease Acta Neurol Scand 93: 18–24

Ihl R, Perisic I, Maurer K, Dierks T (1989) Effect of 3 months treatment with tenilsetamin patients suffering from dementia of Alzheimer type (DAT). J Neural Transm[P-D Sect]1: 84–85

Imani F, Horii Y, Suthanthiran M, Skolnik EY, Makita Z, Sharma V, Sehajpal P, VlassaraH. (1993) Advanced glycosylation endproduct-specific receptors on human and ratT-lymphocytes mediate synthesis of interferon-gamma – role in tissue remodeling.J Exp Med 178: 2165–2172

Irizarry MC, McNamara M, Fedorchak K, Hsiao K, Hyman BT (1997) APP(SW)transgenic mice develop age-related Aâ deposits and neuropil abnormalities, but noneuronal loss in CA1. J Neuropathol Exp Neurol 56: 965–973

Jarrett JT, Lansbury PT Jr (1993) Seeding “one-dimensional crystallization” ofamyloid: a pathogenic mechanism in Alzheimer’s disease and scrapie? Cell 73: 1055–1058

Kaltschmidt B, Uherek M, Volk B, Baeuerle PA, Kaltschmidt C (1997) Transcriptionfactor NFκB is activated in primary neurons by amyloid beta peptides and in neuronssurrounding early plaques from patients with Alzheimer disease. Proc Natl Acad SciUSA 94: 2642–2647

Page 19: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 457

Kimura T, Takamatsu J, Araki K (1995) Are advanced glycation end-products associatedwith amyloidosis in Alzheimer’s disease? Neuroreport 6: 866–868

Kimura T, Takamatzsu J, Ikeda K, Kondo A, Miyakawa T, Horiuchi S (1996) Accumula-tion of advanced glycation end products of the Maillard reaction with age in humanhippocampal neurons. Neurosci Lett 208: 53–56

Kohen R, Fanberstein D, Tirosh O (1997) Reducing equivalents in the aging process.Arch Gerontol Geriatr 24: 103–123

Lander HM, Tauras JM, Ogiste JS, Hori O, Moss RA, Schmidt AM (1997) Activation ofthe receptor for advanced glycation end products triggers a p21(ras)-dependentmitogen-activated protein kinase pathway regulated by oxidant stress. J Biol Chem272: 17810–17814

Ledesma MD, Bonay P, Colaco C, Avila J (1994) Analysis of microtubule-associated protein tau glycation in paired helical filaments. J Biol Chem 269: 21614–21619

Ledesma MD, Bonay P, Avila J (1995) Tau protein from Alzheimer’s disease patients isglycated at its tubulin-binding domain. J Neurochem 65: 1658–1664

Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E, OBrien PC, Palumbo PJ (1997)Risk of dementia among persons with diabetes mellitus – a population-based cohortstudy. Am J Epidemiol 145: 301–308

Lieb K, Fiebich BL, Hüll M, Berger M, Bauer J (1997) Potent inhibition of interleukin-6 expression in a human astrocytoma cell line by tenidap. Cell Tissue Res 288: 251–257

Li JJ, Voisin D, Quiquerez AL, Bouras C (1994) Differential expression of ad-vanced glycation end-products in neurons of different species. Brain Res 641: 285–288

Li JJ, Surini M, Catsicas S, Kawashima E, Bouras C (1995) Age-dependent accumulationof advanced glycosylation end products in human neurons. Neurobiol Aging 16: 69–76

Li JJ, Dickson D, Hof P, Vlassara H (1998) Receptors for advanced glycationendproducts in human brain: Role in brain homeostasis. Mol Med 4: 46–60

Li YM, Dickson DW (1997) Enhanced binding of advanced glycation endproducts(AGE) by the Apoe4 isoform links the mechanism of plaque deposition inAlzheimers disease. Neurosci Lett 226: 155–158

Li YM, Mitsuhashi T, Wojciechowicz D, Shimizu N, Li J, Stitt A, He C, Banerjee D,Vlassara H (1996) Molecular identity and cellular distribution of advanced glycationendproduct receptors: relationship of p60 to OST-48 and p90 to 80K-H membraneproteins. Proc Natl Acad Sci USA 93: 11047–11052

Loo DT, Copani A, Pike CJ, Whittemore ER, Walencewicz AJ, Cotman CW (1993)Apoptosis is induced by beta-amyloid in cultured central nervous system neurons.Proc Natl Acad Sci USA 90: 7951–7955

Lorenzo A, Yankner BA (1996) Amyloid fibril toxicity in Alzheimer’s disease anddiabetes. Ann NY Acad Sci 777: 89–95

Maines MD (1988) Heme oxygenase: function, multiplicity, regulatory mechanisms, andclinical applications. FASEB J 2: 2557–2568

Mandelkow EM, Schweers O, Drewes G, Biernat J, Gustke N, Trinczek B, Mandelkow E(1996) Structure, microtubule interactions, and phosphorylation of tau protein.Ann NY Acad Sci 777: 96–106

Mark RJ, Pang Z, Geddes JW, Uchida K, Mattson MP (1997) Amyloid beta-peptideimpairs glucose transport in hippocampal and cortical neurons: involvement of mem-brane lipid peroxidation. J Neurosci 17: 1046–1054

McGeer EG, McGeer PL (1997) Inflammatory cytokines in the CNS – possible role in thepathogenesis of neurodegenerative disorders and therapeutic implications. CNSDrugs 7: 214–228

McGeer PL, Rogers J, McGeer EG (1994) Neuroimmune mechanisms in Alzheimerdisease pathogenesis. Alzheimer Dis Assoc Disord 8: 149–158

Page 20: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

458 G. Münch et al.

McMillian M, Kong LY, Sawin SM, Wilson B, Das K, Hudson P, Hong JS, Bing G(1995) Selective killing of cholinergic neurons by microglial activation in basalforebrain mixed neuronal/glial cultures. Biochem Biophys Res Commun 215: 572–577

McShea A, Harris PL, Webster KR, Wahl AF, Smith MA (1997) Abnormal expression ofthe cell cycle regulators P16 and CDK4 in Alzheimer’s disease. Am J Pathol 150:1933–1939

McShea A, Wahl AF, Smith MA (1998) Re-entry into the cell cycle: a mechanism forneurodegeneration in Alzheimer’s disease. Med Hypotheses (in press)

Mecocci P, Beal MF, Cecchetti R, Polidori MC, Cherubini A, Chionne F, Avellini L,Romano G, Senin U (1997) Mitochondrial membrane fluidity and oxidative damageto mitochondrial DNA in aged and AD human brain. Mol Chem Neuropathol 31: 53–64

Miyata T, Hori O, Zhang JH, Yan SD, Ferran L, Lida Y, Schmidt AM (1996) Thereceptor for advanced glycation end products (RAGE) is a central mediator of theinteraction of age-beta(2)microglobulin with human mononuclear phagocytes via anoxidant-sensitive pathway – implications for the pathogenesis of dialysis-relatedamyloidosis. J Clin Invest 98: 1088–1094

Monnier VM, Cerami A (1981) Nonenzymatic browning in vivo: possible process foraging of long-lived proteins. Science 211: 491–493

Monnier VM, Sell DR, Nagaraj RH, Miyata S (1991) Mechanisms of protection againstdamage mediated by the Maillard reaction in aging. Gerontology 37: 152–165

Montine TJ, Huang DY, Valentine WM, Amarnath V, Saunders A, Weisgraber KH,Graham DG, Strittmatter WJ (1996a) Crosslinking of apolipoprotein E by productsof lipid peroxidation. J Neuropathol Exp Neurol 55: 202–210

Montine TJ, Amarnath V, Martin ME, Strittmatter WJ, Graham DG (1996b) E-4-hydroxy-2-nonenal is cytotoxic and cross-links cytoskeletal proteins in P19 neuroglialcultures. Am J Pathol 148: 89–93

Münch G, Taneli Y, Schraven E, Schindler U, Schinzel R, Palm D, Riederer P (1994)The cognition-enhancing drug tenilsetam is an inhibitor of protein crosslinking byadvanced glycosylation. J Neural Transm [P-D Sect] 8: 193–208

Münch G, Mayer S, Michaelis J, Hipkiss AR, Schinzel R, Riederer P, Cunningham AM(1997a) Influence of Advanced Glycosylation Endproducts and AGE – inhibitors onnucleation dependent polymerization of â – amyloid peptide. Biochem Biophys Acta[Mol Bas Dis Sect] 1360: 17–29

Münch G, Loske C, Simm A, Schinzel R, Riederer P (1997b) Cytotoxicity of advancedglycation endproducts – protection by antioxidants. Biol Chem 378: S 139

Mullarkey CJ, Edelstein D, Brownlee M (1990) Free radical generation by early glycationproducts: a mechanism for accelerated atherogenesis in diabetes. Biochem BiophysRes Commun 173: 932–939

Namiki M (1988) Chemistry of Maillard reactions: recent studies on the browning reac-tion mechanism and the development of antioxidants and mutagens. Adv Food Res32: 115–184

Novak M, Kabat J, Wischik CM (1993) Molecular characterization of the minimalprotease resistant tau unit of the Alzheimer’s disease paired helical filament. EMBOJ 12: 365–370

Ohkura T, Isse K, Akazawa K, Hamamoto M, Yaoi Y, Hagino N (1994) Evaluation ofestrogen treatment in female patients with dementia of the Alzheimer type. EndocrJ 41: 361–371

Ott A, Stolk RP, Hofman A, Vanharskamp F, Grobbee DE, Breteler MMB (1996)Association of diabetes mellitus and dementia – the Rotterdam study. Diabetologia39: 1392–1397

Parnetti L, Gaiti A, Polidori MC, Brunetti M, Palumbo B, Chionne F, Cadini D, CecchettiR, Senin U (1995) Increased cerebrospinal fluid pyruvate levels in Alzheimersdisease. Neurosci Lett 199: 231–233

Page 21: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 459

Perani D, Grassi F, Sorbi S, Nacmias B, Piacentini S, Piersanti P, Provinciali L, AmaducciL, Fazio F (1997) PET study in subjects from two italian FAD families with APP717Val to Ile mutation. Eur J Neurol 4: 214–220

Praprotnik D, Smith MA, Richey PL, Vinters HV, Perry G (1996) Plasma membranefragility in dystrophic neurites in senile plaques of Alzheimer’s disease: an index ofoxidative stress. Acta Neuropathol 91: 1–5

Pike CJ, Walencewicz-Wasserman AJ, Kosmoski J, Cribbs DH, Glabe CG, Cotman CW(1995) Structure-activity analyses of beta-amyloid peptides – contributions of thebeta-25-35 region to aggregation and neurotoxicity. J Neurochem 64: 253–265

Premkumar DRD, Smith MA, Richey PL, Petersen RB, Castellani R, Kutty RK, WiggertB, Perry G, Kalaria RN (1995) Induction of heme oxygenase-1 mRNA and protein inneocortex and cerebral vessels in Alzheimer’s disease. J Neurochem 65: 1399–1402

Pollard HB, Arispe N, Rojas E (1995) Ion channel hypothesis for Alzheimer amyloidpeptide neurotoxicity. Cell Mol Neurobiol 15: 513–526

Price DL, Sisodia SS (1994) Cellular and molecular biology of Alzheimer’s disease andanimal models. Ann Rev Med 45: 435–446

Rapoport SI, Hatanpaa K, Brady DR, Chandrasekaran K (1996) Brain energy metabo-lism, cognitive function and down-regulated oxidative phosphorylation in Alzheimersdisease. Neurodegeneration 5: 473–476

Richards GA, van Antwerpen VL, Hunter S, Theron AJ, Vandermerwe CA, VanderwaltR, Anderson R (1997) Ageing and cigarette smoking are associated with decreasedglutathione levels in humans. S Afr J Sci 92: 445–447

Roy S, Sen CK, Tritschler HJ, Packer L (1997) Modulation of cellular reducing equivalenthomeostasis by alpha-lipoic acid – mechanisms and implications for diabetes andischemic injury. Biochem Pharmacol 53: 393–399

Saada A, Reichert F, Rotshenker S (1996) Granulocyte macrophage colony stimulatingfactor produced in lesioned peripheral nerves induces the upregulation of cell surfaceexpression of MAC-2 by macrophages and Schwann cells. J Cell Biol 133: 159–167

Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, Grundman M, Woodbury P,Growdon J, Cotman CW, Pfeiffer E, Schneider LS, Thal LJ (1997) A controlled trialof selegiline, alpha-tocopherol, or both as treatment for Alzheimer’s disease. N EnglJ Med 336: 1216–1222

Sanz N, Diez-Fernandez C, Alvarez A, Cascales M (1997) Age-dependent modificationsin rat hepatocyte antioxidant defense systems. J Hepatol 27: 525–534

Sayre LM, Arora PK, Iyer RS, Salomon RG (1993) Pyrrole formation from 4-hydroxynonenal and primary amines. Chem Res Toxicol 6: 19–22

Sayre LM, Zelasko DA, Harris PLR, Perry G, Salomon RG, Smith MA (1997) 4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased inAlzheimers disease. J Neurochem 68: 2092–2097

Schmidt AM, Yan SD, Brett J, Mora R, Nowygrod R, Stern D (1993) Regulationof human mononuclear phagocyte migration by cell surface-binding proteins foradvanced glycation end products. J Clin Invest 91: 2155–2168

Schmidt AM, Hori O, Brett J, Yan SD, Wautier JL, Stern D (1994) Cellular receptorsfor advanced glycation end products. Implications for induction of oxidant stressand cellular dysfunction in the pathogenesis of vascular lesions. Arterioscler Thromb14: 1521–1528

Schmidt AM, Hori O, Cao R, Yan SD, Brett J, Wautier JL, Ogaea S, Kuwabara K,Matsumoto M, Stern D (1996) RAGE, a novel cellular receptor for advancedglycation endproducts. Diabetes 45: 77–80

Schweers O, Mandelkow EM, Biernat J, Mandelkow E (1995) Oxidation of cysteine-322in the repeat domain of microtubule-associated protein tau controls the in vitroassembly of paired helical filaments. Proc Natl Acad Sci USA 92: 8463–8467

Sheu KFR, Cooper AJL, Koike K, Koike M, Lindsay JG, Blass JP (1994) Abnormalityof the alpha-ketoglutarate dehydrogenase complex in fibroblasts from familialAlzheimers disease. Ann Neurol 35: 312–318

Page 22: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

460 G. Münch et al.

Shoda H, Miyata S, Liu BF, Yamada H, Ohara T, Suzuki K, Oimomi M, Kasuga M (1997)Inhibitory effects of tenilsetam on the Maillard reaction. Endocrinology 138: 1886–1892

Simm A, Münch G, Seif F, Schenk O, Heidland A, Richter H, Vamvakas S, Schinzel R(1997) Advanced glycation endproducts activate the MAP kinase pathway in thetubulus cell line LLC-PK1. FEBS Lett 410: 481–484

Smedsrod B, Melkko J, Araki N, Sano H, Horiuchi S (1997) Advanced glycation endproducts are eliminated by scavenger-receptor-mediated endocytosis in hepaticsinusoidal Kupffer and endothelial cells. Biochem J 322: 567–573

Smith MA, Kutty RK, Richey PL, Yan SD, Stern D, Chader GJ, Wiggert B, Petersen RB,Perry G (1994a) Heme oxygenase-1 is associated with the neurofibrillary pathology ofAlzheimers disease. Am J Pathol 145: 42–47

Smith MA, Richey PL, Taneda S, Kutty RK, Sayre LM, Monnier VM, Perry G (1994b)Advanced Maillard reaction end products, free radicals, and protein oxidation inAlzheimers disease. Ann NY Acad Sci 738: 447–454

Smith MA, Taneda S, Richey PL, Miyata S, Yan SD, Stern D, Sayre LM, Monnier VM,Perry G (1994c) Advanced Maillard reaction end products are associated withAlzheimer disase pathology. Proc Natl Acad Sci USA 91: 5710–5714

Smith MA, Sayre LM, Monnier VM, Perry G (1995) Radical AGEing in Alzheimer’sdisease. Trends Neurosci 18: 172–176

Smith MA, Perry G, Richey PL, Sayre LM, Anderson VE, Beal MF, Kowall N (1996a)Oxidative damage in Alzheimers. Nature 382: 120–121

Smith MA, Sayre LM, Perry G (1996b) Diabetes mellitus and Alzheimer’s disease:glycation as a biochemical link. Diabetologia 39: 247

Smith MA, Harris PLR, Sayre LM, Perry G (1997) Iron accumulation in Alzheimerdisease is a source of redox-generated free radicals. Proc Natl Acad Sci USA 94:9866–9868

Vitek MP, Bhattacharya K, Glendening JM, Stopa E, Vlassara H, Bucala R, Manogue K,Cerami A (1994) Advanced glycation end products contribute to amyloidosis inAlzheimer disease. Proc Natl Acad Sci USA 91: 4766–4770

Vlassara H, Brownlee M, Manogue K, Dinarello C, Pasagian A (1988) Cachectin/TNFand IL-1 induced by glucose-modified proteins: role in normal tissue remodeling.Science 240: 1546–1550

Vlassara H, Moldawer L, Chan B (1989) Macrophage/monocyte receptor fornonenzymatically glycosylated proteins is upregulated by cachectin/tumor necrosisfactor. J Clin Invest 84: 1831–1820

Vlassara H, Li YM, Imani F, Wojciechowicz D, Yang Z, Liu FT, Cerami A (1995)Identification of galectin-3 as a high-affinity binding protein for advanced glycationend products (AGE): a new member of the AGE-receptor complex. Mol Med 1: 634–646

Wautier JL, Wautier MP, Schmidt AM, Anderson GM, Hori O, Zoukourian C, CapronL, Chappey O, Yan SD, Brett J (1994) Advanced glycation end products (AGEs) onthe surface of diabetic erythrocytes bind to the vessel wall via a specific receptorinducing oxidant stress in the vasculature: a link between surface-associated AGEsand diabetic complications. Proc Natl Acad Sci USA 91: 7742–7746

Wells-Knecht KJ, Zyzak DV, Litchfield JE, Thorpe SR, Baynes JW (1995a) Mechanismof autoxidative glycosylation – identification of glyoxal and arabinose as inter-mediates in the autoxidative modification of proteins by glucose. Biochem 34: 3702–3709

Wells-Knecht MC, Thorpe SR, Baynes JW (1995b) Pathways of formation ofglycoxidation products during glycation of collagen. Biochem 34: 15134–15141

Yan SD, Chen X, Schmidt AM, Brett J, Godman G, Zou YS, Scott CW, Caputo C,Frappier T, Smith MA, Perry G, Yen SH, Stern D (1994) Glycated tau protein inAlzheimer disease: a mechanism for induction of oxidant stress. Proc Natl Acad SciUSA 91: 7787–7791

Page 23: Alzheimer's disease – synergistic effects of glucose deficit, oxidative stress and advanced glycation endproducts

Alzheimer’s disease – synergistic effects 461

Yan SD, Yan SF, Chen X, Fu J, Chen M, Kuppusamy P, Smith MA, Perry G, GodmanGC, Nawroth P, Stern D (1995) Non-enzymatically glycated tau in Alzheimer’sdisease induces neuronal oxidant stress resulting in cytokine gene expression andrelease of amyloid beta-peptide. Nature Med 1: 693–9

Yan SD, Chen X, Fu J, Chen M, Zhu HJ, Roher A, Slattery T, Zhao L, Nagashima M,Morser J, Migheli A, Nawroth P, Stern D, Schmidt AM (1996) RAGE and amyloid-beta peptide neurotoxicity in Alzheimers disease. Nature 382: 685–691

Yang Z, Makita Z, Horii Y, Brunelle S, Cerami A, Sehajpal P, Suthanthiran M, VlassaraH (1991) Two novel rat liver membrane proteins that bind advanced glycosylationendproducts: relationship to macrophage receptor for glucose-modified proteins.J Exp Med 174: 515–524

Zhang ZY, Rydel RE, Drzewiecki GJ, Fuson K, Wright S, Wogulis M, Audia JE, May PC,Hyslop PA (1996) Amyloid beta-mediated oxidative and metabolic stress in ratcortical neurons – no direct evidence for a role for H2O2 generation. J Neurochem67: 1595–1606

Authors’ address: Dr. G. Münch, Physiological Chemistry I, Biocenter Am Hubland,D-97074 Würzburg, Federal Republic of Germany.