eISSN: 1509-572x
ISSN: 1641-4640
Folia Neuropathologica
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2005
vol. 43
 
Share:
Share:

Original article
Alzheimer\'s disease related peptides affected cholinergic receptor mediated poly(ADP-ribose) polymerase activity in the hippocampus

Agata Adamczyk
,
Henryk Jęśko
,
Robert Piotr Strosznajder

Folia Neuropathol 2005; 43 (3): 139-142
Online publish date: 2005/09/30
Article file
- Alzheimer.pdf  [0.09 MB]
Get citation
 
 
Communicating author:
Agata Adamczyk, Department of Cellular Signaling, Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego St, 02-106 Warsaw, Poland, tel. /fax +48 22 608 65 72, e-mail: agatazambrzycka@hotmail.com

Introduction
Our previous study presented that activation of the muscarinic cholinergic receptor (mChR) by non hydrolysable analog of acetylcholine, carbachol and activator of G protein, GTP(g)S significantly enhanced inositol-1,4,5-triphosphate (IP3) and diacylglycerol (DAG) formation and that this signaling events are significantly altered by amyloid beta peptide (Aβ) [27]. In this study, the mChR evoked signals were investigated on the level of DNA-bound enzyme poly(ADP-ribose) polymerase (PARP-1, EC 2.4.2.30). The recent data suggested that PARP-1 is the new nuclear target for signal transduction processes evoked by receptor(s) stimulation or membrane depolarization [12,21,23]. Activated PARP-1 cleaves NAD into nicotinamide and forms long and branched polymers of ADP-ribose on over forty nuclear proteins and
PARP-1 itself. Poly (ADP-ribosyl) ation has been implicated in the regulation of a diverse array of cellular processes ranging from DNA repair, chromatin organization, transcription, to replication [2,7,9,15,21,25]. However, oxidative stress-induced DNA strand breaks that stimulates PARP-1 lead to bNAD+ and ATP depletion and to necrotic or apoptotic processes [10,13,26]. The pathophysiological significance of PARP-1 over-activation is suggested in brain ischemia, diabetes, inflammation and cancer [4,11,21]. Increased expression of this enzyme and accumulation of poly(ADP-ribose) was observed in Alzheimer's and Parkinson's brain [14,17,19]. Our resent data indicated the involvement of mChR
in the regulation of PARP-1 activity through IP3 receptor pathway [23]. In the present study, we investigated the effect of Alzheimer's disease (AD) related Aβ and NAC peptide on mChR-dependent signal transduction to PARP-1.

Material and methods
Animals

Male Wistar rats, 4-month-old (250-300 g), were supplied from the Animal Breeding House of the Medical Research Centre (Warsaw). The Institutional Ethics Committee accepted the research project.

Materials
Adenine[C14]NAD (sp. activity: 252 mCi/mmol), was obtained from Amersham, Buckinghamshire, UK, Aβ 25-35, carbachol, GTPgS, and the all other reagents were from Sigma Chemical Co. (St. Louis, MO, U.S.A.).

Preparation of Aβ and NAC peptides
Aβ 25-35 was dissolved in bidistilled deionized (BDD) water at 2.5 mM concentration and incubated at room temperature to obtain the aggregated form as described previously [20]. Aβ was used at 25 µM final concentration. NAC peptide was stored lyophilized, and 100 µM stock solution was prepared in BDD water and then used at final 10 µM concentration. To obtain the aggregated form NAC was incubated for 3 days at 37°C.

Preparation of brain slices
Animals were killed by decapitation, the brain was quickly removed, hippocampi were dissected and cross-chopped into slices (350x350 µm) using a McIlwain tissue chopper. The slices were placed in ice-cold Krebs buffer (in mM: NaCl 124, KCl 5, MgSO4 1.2, KH2PO4 1.2, NaHCO3 26 and glucose 10) equilibrated with 5% CO2 in 95% O2 to maintain a pH of 7.4 and pre-incubated for 30 min at 37°C in Krebs buffer. Then, CaCl2 was added to final 2 mM concentration and incubation was prolonged for 30 min at 37°C in the presence or absence of the following compounds: Aβ 25-35 (25 µM), NAC (10 µM), carbachol (1 mM) together with GTP(g)S (100 µM), TMB-8 (10 µM). After incubation, the slices were homogenized in glass-glass homogenizer and used for determination of the PARP-1 activity.

Determination of the PARP-1 activity
The PARP-1 activity was assayed as described previously [22]. The total reaction mixture in the final volume of 100 µl contained 400µM [adenine-14C]NAD as a substrate (2x105 d.p.m., sp. activity: 252 mCi/mmol), 100 mM Tris-HCl buffer pH 8.0, 10 mM MgCl2, 1mM dithiothreitol (DTT) and 200 µg of protein. The reaction was carried out for 1 min at 37°C and stopped with 800 µl of ice-cold 25% trichloroacetic acid (TCA). The precipitate was collected on Whatman GF/B filters, washed five times with ice-cold 5% TCA and processed for determination of radioactivity in Bray's scintillation fluid using scintilator counter Wallac 1409 LKB.
Results
In the present study, we examined the role of mChR on PARP-1 activation in the brain cortex and hippocampus. Moreover, the effect of both AD related peptide Aβ and NAC on this receptor mediated signaling to PARP-1 was evaluated. The data showed that carbachol at 1mM concentration together with GTP(g)S at 100 µM significantly enhanced the PARP-1 activity by about 100% in the hippocampus with no effect on this enzyme in the brain cortex (Fig. 1). Inhibition of IP3 receptor through TMB-8 (10 µM) decreased PARP-1 activation evoked by carbachol/GTP(g)S in the hippocampus (Table I). MChR stimulation by carbachol/GTP(g)S had no effect on free radicals generation and macromolecules oxidation (data not shown). In addition, this receptor stimulation had no effect on the PARP-1 activity in the presence of Aβ and NAC peptide (Fig. 2). However, both peptides, Aβ and NAC significantly stimulated this enzyme by 88% and 23%, respectively by free radicals evoked DNA strand breaks (Fig. 2).

Discussion
The major finding of this study is that AD related peptide Aβ and NAC disturbed phosphoinositide signaling to PARP-1 in the hippocampus. These and our recent results presented PARP-1 as a nuclear target for mChR pathway [23] are in agreement with the previous data of Homburg et al [12]. They showed that depolarisation of neurons activated PARP via
IP3-induced Ca2+ signaling. In addition, it was shown that N-methyl-D-aspartate (NMDA) receptors stimulation could mediate PARP-1 activation [18,22]. Moreover, the present data indicated that Aβ and NAC peptide enhanced basal PARP-1 activity. These results supported our earlier data that both peptides induced reactive oxygen species (ROS) generation and DNA degradation [1]. It is well documented that PARP-1 is the earliest and the most sensitive indicator of DNA strand breaks evoked by oxidative stress [8,28]. Over-activation of PARP-1 leads to cellular bNAD+ depletion, apoptotic inducing factor (AIF) release and cell death [13,26]. Aβ together with NAC peptide is a major component of senile plaques in AD and through ROS generation could participate in neuronal cell death [3,5,6,16,24]. Our previous results showed that mChR-dependent signal transduction in the rat brain is damaged by Aβ peptide that significantly inhibited phosphatidylinositol-4,5-bisphosphate phospholipase C (PIP2-PLC) activity through ROS formation and membrane lipids peroxidation [20,27]. The present data indicated that Aβ and NAC peptide decreased PARP-1 activation evoked by mChR stimulation. We therefore consider that both peptides through the free radicals mechanism inhibited PIP2-PLC activity and IP3 liberation and disturbed signal transduction from mChR to PARP-1.

Acknowledgements
This work was supported by the Grant PBZ-MIN-001/P05/16 and by statutory budget of the Polish Academy of Science Medical Research Center (Theme No 1).

References
1. Adamczyk A, Czapski GA, Jęśko H, Strosznajder RP. Non-Aβ component of Alzheimer's disease amyloid and amyloid beta peptides evoked poly(ADP-ribose) polymerase-dependent release of apoptosis-inducing factor from rat brain mitochondria. J Physiol Pharmacol 2005; 56 (Supp. 2): 5-13.
2. Ame JC, Spenlehauer C, de Murcia G. The PARP superfamily. Bioessays 2004; 26: 882-893.
3. Behl C, Davis JB, Lesley R, Schubert D. Hydrogen peroxide mediates amyloid beta protein toxicity. Cell 1994; 77: 817-827.
4. Besson VC, Zsengeller Z, Plotkine M, Szabo C, Marchand-Verrecchia C. Beneficial effects of PJ34 and INO-1001, two novel water-soluble poly(ADP-ribose) polymerase inhibitors, on the consequences of traumatic brain injury in rat. Brain Res 2005; 1041: 149-156.
5. Butterfield DA, Boyd-Kimball D. Amyloid beta-peptide (1-42) contributes to the oxidative stress and neurodegeneration found in Alzheimer disease brain. Brain Pathol 2004; 14: 426-432.
6. Butterfield DA, Boyd-Kimball D. The critical role of methionine 35 in Alzheimer's amyloid beta-peptide (1-42) -induced oxidative stress and neurotoxicity. Biochim Biophys Acta 2005; 703: 149-156.
7. Chang WJ, Alvarez-Gonzalez R. The sequence-specific DNA binding of NF-kappa B is reversibly regulated by the automodification reaction of poly(ADP-ribose) polymerase 1. J Biol Chem 2001; 276: 47664-47670.
8. de Murcia G, Menissier de Murcia J. Poly(ADP-ribose) polymerase: a molecular nick-sensor. Trends Biochem Sci 1994; 19: 172-176.
9. Decker P, Muller S. Modulating poly(ADP-ribose) polymerase activity: potential for the prevention and therapy of pathogenic situations involving DNA damage and oxidative stress. Curr Pharm Biotechnol 2002; 3: 275-283.
10. Erdelyi K, Bakondi E, Gergely P, Szabo C, Virag L. Pathophysiologic role of oxidative stress-induced poly(ADP-ribose) polymerase-1 activation: focus on cell death and transcriptional regulation. Cell Mol Life Sci 2005; 62: 751-759.
11. Graziani G, Szabo C. Clinical perspectives of PARP inhibitors. Pharmacol Res 2005; 52: 109-118.
12. Homburg S, Visochek L, Moran N, Dantzer F, Priel E, Asculai E, Schwartz D, Rotter V, Dekel N, Cohen-Armon M. A fast signal-induced activation of Poly(ADP-ribose) polymerase: a novel downstream target of phospholipase c. J Cell Biol 2000; 150: 293-307.
13. Hong SJ, Dawson TM, Dawson VL. Nuclear and mitochondrial conversations in cell death: PARP-1 and AIF signaling. Trends Pharmacol Sci 2004; 25: 259-264.
14. Iwashita A, Yamazaki S, Mihara K, Hattori K, Yamamoto H, Ishida J, Matsuoka N, Mutoh S. Neuroprotective effects of a novel poly (ADP-ribose) polymerase-1 inhibitor, 2-[3-[4-(4-chlorophenyl)-1-piperazinyl] propyl]-4(3H)-quinazolinone (FR255595), in an in vitro model of cell death and in mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson's disease. J Pharmacol Exp Ther 2004; 309: 1067-1078.
15. Jeggo PA. DNA repair: PARP – another guardian angel? Curr Biol 1998; 8: R49-51.
16. Keil U, Bonert A, Marques CA, Scherping I, Weyermann J, Strosznajder JB, Müller-Spahn F, Haass C, Czech C, Pradier L, Müller WE, Eckert A. Amyloid beta-induced changes in nitric oxide production and mitochondrial activity lead to apoptosis. J Biol Chem 2004; 279: 50310-50320.
17. Love S, Barber R, Wilcock GK. Increased poly (ADP-ribosyl) ation of nuclear proteins in Alzheimer's disease. Brain 1999; 122 (Pt 2): 247-253.
18. Pieper AA, Verma A, Zhang J, Snyder SH. Poly(ADP-ribose) polymerase, nitric oxide and cell death. Trends Pharmacol Sci 1999; 20: 171-181.
19. Soos J, Engelhardt JI, Siklos L, Havas L, Majtenyi K. The expression of PARP, NF-kappa B and parvalbumin is increased in Parkinson disease. Neuroreport 2004; 15: 1715-1718.
20. Strosznajder JB, Zambrzycka A, Kacprzak MD, Strosznajder RP. Amyloid beta peptide 25-35 modulates hydrolysis of phosphoinositides by membrane phospholipase (s) C of adult brain cortex. J Mol Neurosci 1999; 12: 101-109.
21. Strosznajder RP, Jesko H, Zambrzycka A. Poly(ADP-ribose) polymerase: the nuclear target in signal transduction and its role in brain ischemia-reperfusion injury. Mol Neurobiol 2005; 31: 149-68.
22. Strosznajder JB, Jesko H, Strosznajder RP. Effect of amyloid beta peptide on poly(ADP-ribose) polymerase activity in adult and aged rat hippocampus. Acta Biochim Pol 2000; 47: 847-854.
23. Strosznajder RP, Jesko H, Adamczyk A. Poly(ADP-ribose) polymerase-1 is the novel nuclear target for cholinergic receptor signaling in hippocampus. J Physiol Pharmacol 2005; (in press).
24. Tanaka S, Takehashi M, Matoh N, Iida S, Suzuki T, Futaki S, Hamada H, Masliah E, Sugiura Y, Ueda K. Generation of reactive oxygen species and activation of NF-kappaB by non-Abeta component of Alzheimer's disease amyloid. J Neurochem 2002; 82: 305-315.
25. Wesierska-Gadek J, Wojciechowski J, Schmid G. Phosphorylation regulates the interaction and complex formation between wt p53 protein and PARP-1. J Cell Biochem 2003; 89: 1260-1284.
26. Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, Federoff HJ, Poirier GG, Dawson TM, Dawson VL. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 2002; 297: 259-263.
27. Zambrzycka A, Strosznajder RP, Strosznajder JB. Aggregated beta amyloid peptide 1-40 decreases Ca2+ – and cholinergic receptor-mediated phosphoinositide degradation by alteration of membrane and cytosolic phospholipase C in brain cortex. Neurochem Res 2000; 25: 189-196.
28. Zhang J, Pieper A, Snyder SH. Poly (ADP-ribose) synthetase activation: an early indicator of neurotoxic DNA damage. J Neurochem 1995; 65: 1411-1414.
Copyright: © 2005 Mossakowski Medical Research Centre Polish Academy of Sciences and the Polish Association of Neuropathologists. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.