14.01.2013 Views

ZMBH J.Bericht 2000 - Zentrum für Molekulare Biologie der ...

ZMBH J.Bericht 2000 - Zentrum für Molekulare Biologie der ...

ZMBH J.Bericht 2000 - Zentrum für Molekulare Biologie der ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

APP catalyzed the reduction of H 2 O 2 and oxidation of<br />

Cu(I) to Cu(II) in a „peroxidative“ reaction in vitro.<br />

The resulting bound copper-hydroxyl radical intermediate<br />

[APP-Cu(II) (•OH)] then likely participated in<br />

a Fenton type of reaction with radical formation as a<br />

prerequisite for APP-Cu(I) complex degradation. Evidence<br />

from two observations suggests that the reaction<br />

takes place in two phases. Bathocuproine, a trapping<br />

agent for Cu(I), abolished the initial fragmentation<br />

of APP, and chelation of Cu(II) by DTPA (diethylenetriaminepentaacetic<br />

acid) interrupted the reaction<br />

cascade induced by H 2 O 2 at later stages. WT cortical,<br />

cerebellar and hippocampal neurons were significantly<br />

more susceptible than their respective APP 0/0 neurons<br />

to toxicity induced by physiological concentrations<br />

of copper but not by zinc or iron. There was no difference<br />

in copper toxicity between APLP2 0/0 and WT<br />

neurons, demonstrating specificity for APP-associated<br />

copper toxicity. Treatment of neuronal cultures with a<br />

peptide corresponding to the human APP copper binding<br />

domain (APP142-166) potentiated copper but not<br />

iron or zinc toxicity. Incubation of APP142-166 with<br />

low density lipoprotein (LDL) and copper resulted<br />

in significantly increased lipid peroxidation compared<br />

to copper and LDL alone. Substitution of the<br />

copper co-ordinating histidine residues with asparagines<br />

(APP142-166 H147N, H149N, H151N ) abrogated the<br />

toxic effects. A peptide corresponding to the zinc binding<br />

domain (APP181-208) failed to induce copper or<br />

zinc toxicity in neuronal cultures. These data support a<br />

role for the APP copper binding domain in APP-mediated<br />

Cu(I) generation and toxicity in primary neurons.<br />

To test the hypothesis that biometals such as copper<br />

can contribute to the amyloid pathology in AD we are<br />

currently investigating the dietary exposure of copper<br />

and its chelators to APP transgenic mice and to APP<br />

42<br />

knock-out mice. The effects of dietary supplementation<br />

is studied by ICP-MS (inductively coupled plasma<br />

mass-spectrometry) to determine the levels of Cu, Zn<br />

and Fe. The AD pathology is analyzed by conventional<br />

methods. We expect that Cu has an effect on the<br />

AD pathology and metal-chelation can delay amyloid<br />

Aß deposition in APP transgenic mice.<br />

II. The role of copper and zinc in the normal<br />

function of the amyloid precursor protein<br />

(APP)<br />

C. Schmidt, A. Simons, M. Strauss, A. Schlicksupp,<br />

G. Multhaup (in collaboration with D.<br />

Ha-Hao, R. Cappai, A. White, C.L. Masters and<br />

K. Beyreuther)<br />

The expression of APP and APLP2 in the brain<br />

suggests they could have an important direct or indirect<br />

role in neuronal metal homeostasis. In APP and<br />

APLP2 knockout mice copper levels were significantly<br />

elevated in both APP 0/0 and APLP2 0/0 cerebral<br />

cortex (40% and 16%, respectively) and liver (80% and<br />

36%, respectively) compared with matched wild-type<br />

(WT) mice. These findings indicate APP and APLP2<br />

expression specifically modulates copper homeostasis<br />

in the liver and cerebral cortex, the latter being a<br />

region of the brain particularly involved in AD. Perturbations<br />

to APP metabolism and in particular, its secretion<br />

or release from neurons may alter copper homeostasis<br />

and explain a disturbed metal-ion homeostasis<br />

observed in AD.<br />

Zinc up to concentrations of 50µM or the presence of<br />

1,10-phenanthroline specifically increased the level of<br />

secreted APP in APP transfected CHO-K1 cells. By<br />

contrast, the level of secreted APP in copper-resistant<br />

CHO-CUR3 cells remained unaffected. APP holoprotein<br />

increased dramatically in CHO-CUR3 cells com-<br />

pared with CHO-K1 cells. The large decrease of Aß<br />

release seen in both cell lines at elevated extracellular<br />

zinc levels was due to specific inhibition of secretion.<br />

These results indicate that a disturbed zinc-homeostasis<br />

may be an important factor influencing APP production,<br />

transport and processing.<br />

Figure 3: The α- and β-secretase protease activities cleave<br />

APP within its ectodomain. The remaining membranebound<br />

C-terminal fragment p3CT (after α-secretase cleavage<br />

of APP) is further cleaved by γ-secretase in the middle<br />

of the putative transmembrane domain, yielding p3 (α- and<br />

γ-secretase activities). Amyloid Aβ is produced by β- and<br />

γ-secretase activities; CuBD-I, copper binding domain 1.<br />

Adding copper to APP-transfected CHO cells greatly<br />

reduced the levels of ß-amyloid (Aß) peptide in both<br />

parental CHO-K1 and in copper resistant CHO-CUR3<br />

cells which have lower intracellular copper levels.<br />

Copper also caused an increase in the secretion of the<br />

APP ectodomain indicating that the large decrease in<br />

Aß release was not due to a general inhibition in protein<br />

secretion. There was an increase in intracellular<br />

full-length APP levels which paralleled the decrease<br />

in Aß generation suggesting the existence of two distinct<br />

regulating mechanisms, one acting on Aß production<br />

and the other on APP synthesis. Thus, our<br />

findings suggest that copper or copper agonists might<br />

be useful tools to discover novel targets for anti-<br />

Alzheimer drugs since copper promoted the non-amyloidogenic<br />

pathway of APP.<br />

Our current un<strong>der</strong>standing is that copper and/or zinc<br />

binding is central to the normal cellular function of<br />

APP. We therefore want to identify agonists of APP<br />

ligand binding sites (e.g. of the copper-binding site)<br />

that are able to inhibit amyloidogenic proteolytic processing<br />

of APP. Lead compounds will be discovered<br />

followed by a further screen for small APP ligands.<br />

Our research program combines biochemical, immunohistochemical<br />

and cellular approaches to start a<br />

risk-benefit analysis of divalent metal binding agonists<br />

of APP in vitro.<br />

III. Dimerization and stability of APP isoforms:<br />

influence of APP ligands on relative<br />

stability and metabolism<br />

M. Strauss, C. Schmidt, A. Simons, A. Schlicksupp,<br />

C. Elle, G. Multhaup (in collaboration with<br />

C. Haass, C.L. Masters and K. Beyreuther)<br />

The highly conserved nature and tissue specificity of<br />

the eight APP isoforms provide circumstantial evidence<br />

that functional differences among isoforms may<br />

exist in vitro and in vivo. When APP and Aß are central<br />

to AD pathogenesis, then molecules which influence<br />

the conformation and the stability of APP isoforms<br />

and that interact with specific APP forms could<br />

differentially alter their metabolism/activity and thus<br />

represent risk factors for AD.<br />

For example, heparin and Zn(II) were found to augment<br />

the ability of full-length and secreted KPI-APP to<br />

inhibit FXIa. In contrast, both compounds heparin and<br />

43

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!