Skip to main content

Advertisement

Log in

Site-Activated Chelators Derived from Anti-Parkinson Drug Rasagiline as a Potential Safer and More Effective Approach to the Treatment of Alzheimer’s Disease

  • ORIGINAL PAPER
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

chelators can modulate β-amyloid accumulation, protect against tau hyperphosphorylation, and block metal-related oxidative stress, and thereby hold considerable promise as effective anti-AD drugs. At present, a growing interest is focusing on increasing the efficacy and targeting of chelators through drug design. To this end, we have developed a new class of multifunctional prochelators from three FDA- approved drugs rasagiline, rivastigmine, and donepezil or tacrine. HLA20 A was designed by merging the important pharmacophores of rasagiline, rivastigmine, and donepezil into our newly developed multifunctional chelator HLA20. M30D was constructed using the key pharmacophoric moieties from rasagiline, rivastigmine, and tacrine. Experiments showed that both compounds possess potent anti-acetylcholinesterase (AChE) activity in vitro with weak inhibition of butyrylcholinesterase (BuChE), and without significant metal-binding activity. M30D was found also to be a highly potent MAO A inhibitor with moderate inhibition of MAO B in vitro. Both HLA20 and M30D can be activated by inhibition of AChE to release active chelators HLA20 and M30, respectively. HLA20 and M30 have been shown to be able to modulate amyloid precursor protein regulation and beta-amyloid reduction, suppress oxidative stress, and passivate excess metal ions (Fe, Cu, and Zn). Compared with the activated chelator HLA20 or M30, both HLA20A and M30D exhibited lower cytotoxicity in SH-SY5Y neuroblastoma cells, substantiating the prochelator strategy for minimizing toxicity associated with poor targeted chelators.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Biran Y, Masters CL, Barnham KJ et al (2009) Pharmacotherapeutic targets in Alzheimer’s disease. J Cell Mol Med 13:61–86

    Article  CAS  PubMed  Google Scholar 

  2. Mattson MP (2004) Pathways towards and away from Alzheimer’s disease. Nature 430:631–639

    Article  CAS  PubMed  Google Scholar 

  3. Barten DM, Albright CF (2008) Therapeutic strategies for Alzheimer’s disease. Mol Neurobiol 37:171–186

    Article  CAS  PubMed  Google Scholar 

  4. Lovell MA, Robertson JD, Teesdale WJ et al (1998) Copper, iron and zinc in Alzheimer’s disease senile plaques. J Neurol Sci 158:47–52

    Article  CAS  PubMed  Google Scholar 

  5. Barnham KJ, Bush AI (2008) Metals in Alzheimer’s and Parkinson’s diseases. Curr Opin Chem Biol 12:222–228

    Article  CAS  PubMed  Google Scholar 

  6. Bandyopadhyay S, Huang X, Cho H et al (2006) Metal specificity of an iron-responsive element in Alzheimer’s APP mRNA 5’untranslated region, tolerance of SH-SY5Y and H4 neural cells to desferrioxamine, clioquinol, VK-28, and a piperazine chelator. J Neural Transm Suppl 71:237–247

    Article  CAS  PubMed  Google Scholar 

  7. Rogers JT, Randall JD, Cahill CM et al (2002) An iron-responsive element type II in the 5′-untranslated region of the Alzheimer’s amyloid precursor protein transcript. J Biol Chem 277:45518–45528

    Article  CAS  PubMed  Google Scholar 

  8. Sayre LM, Perry G, Smith MA (2008) Oxidative stress and neurotoxicity. Chem Res Toxicol 21:172–188

    Article  PubMed  Google Scholar 

  9. Price KA, Crouch PJ, White AR (2007) Therapeutic treatment of Alzheimer’s disease using metal complexing agents. Recent Pat CNS Drug Discov 2:180–187

    Article  CAS  PubMed  Google Scholar 

  10. Adlard PA, Cherny RA, Finkelstein DI et al (2008) Rapid restoration of cognition in Alzheimer’s transgenic mice with 8-hydroxy quinoline analogs is associated with decreased interstitial Abeta. Neuron 59:43–55

    Article  CAS  PubMed  Google Scholar 

  11. Crapper McLachlan DR, Dalton AJ, Kruck TP et al (1991) Intramuscular desferrioxamine in patients with Alzheimer’s disease. Lancet 337:1304–1308

    Article  CAS  PubMed  Google Scholar 

  12. Kaur D, Yantiri F, Rajagopalan S et al (2003) Genetic or pharmacological iron chelation prevents MPTP-induced neurotoxicity in vivo: a novel therapy for Parkinson’s disease. Neuron 37:899–909

    Article  CAS  PubMed  Google Scholar 

  13. Arasaki K, Nakanishi T (1989) Selective neurotoxicity of clioquinol on the function of the posterior column nuclei. Neurosci Lett 107:85–88

    Article  CAS  PubMed  Google Scholar 

  14. Porter JB, Huehns ER (1989) The toxic effects of desferrioxamine. Baillieres Clin Haematol 2:459–474

    Article  CAS  PubMed  Google Scholar 

  15. Zheng H, Youdim MB, Fridkin M (2009) Site-activated multifunctional chelator with acetylcholinesterase and neuroprotective-neurorestorative moieties for Alzheimer’s therapy. J Med Chem 52:4095–4098

    Article  CAS  PubMed  Google Scholar 

  16. Chen JJ, Swope DM, Dashtipour K (2007) Comprehensive review of rasagiline, a second-generation monoamine oxidase inhibitor, for the treatment of Parkinson’s disease. Clin Ther 29:1825–1849

    Article  CAS  PubMed  Google Scholar 

  17. Munoz-Torrero D (2008) Acetylcholinesterase inhibitors as disease-modifying therapies for Alzheimer’s disease. Curr Med Chem 15:2433–2455

    Article  CAS  PubMed  Google Scholar 

  18. Zhang HY, Yan H, Tang XC (2008) Non-cholinergic effects of huperzine A: beyond inhibition of acetylcholinesterase. Cell Mol Neurobiol 28:173–183

    Article  PubMed  Google Scholar 

  19. Amit T, Avramovich-Tirosh Y, Youdim MB et al (2008) Targeting multiple Alzheimer’s disease etiologies with multimodal neuroprotective and neurorestorative iron chelators. FASEB J 22:1296–1305

    Article  CAS  PubMed  Google Scholar 

  20. Zheng H, Gal S, Weiner LM et al (2005) Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases: in vitro studies on antioxidant activity, prevention of lipid peroxide formation and monoamine oxidase inhibition. J Neurochem 95:68–78

    Article  CAS  PubMed  Google Scholar 

  21. Zheng H, Weiner LM, Bar-Am O et al (2005) Design, synthesis, and evaluation of novel bifunctional iron-chelators as potential agents for neuroprotection in Alzheimer’s, Parkinson’s, and other neurodegenerative diseases. Bioorg Med Chem 13:773–783

    Article  CAS  PubMed  Google Scholar 

  22. Youdim MB, Edmondson D, Tipton KF (2006) The therapeutic potential of monoamine oxidase inhibitors. Nat Rev Neurosci 7:295–309

    Article  CAS  PubMed  Google Scholar 

  23. Saura J, Luque JM, Cesura AM et al (1994) Increased monoamine oxidase B activity in plaque-associated astrocytes of Alzheimer brains revealed by quantitative enzyme radioautography. Neuroscience 62:15–30

    Article  CAS  PubMed  Google Scholar 

  24. Reinikainen KJ, Soininen H, Riekkinen PJ (1990) Neurotransmitter changes in Alzheimer’s disease: implications to diagnostics and therapy. J Neurosci Res 27:576–586

    Article  CAS  PubMed  Google Scholar 

  25. Yogev-Falach M, Bar-Am O, Amit T et al (2006) A multifunctional, neuroprotective drug, ladostigil (TV3326), regulates holo-APP translation and processing. FASEB J 20:2177–2179

    Article  CAS  PubMed  Google Scholar 

  26. Zheng H, Youdim MB, Fridkin M (2010) Site-activated chelators targeting acetylcholinesterase and monoamine oxidase for Alzheimer’s therapy. ACS Chem Biol 5:603–610

    Article  CAS  PubMed  Google Scholar 

  27. Groner E, Ashani Y, Schorer-Apelbaum D et al (2007) The kinetics of inhibition of human acetylcholinesterase and butyrylcholinesterase by two series of novel carbamates. Mol Pharmacol 71:1610–1617

    Article  CAS  PubMed  Google Scholar 

  28. Maurer T, Fung HL (2000) Comparison of methods for analyzing kinetic data from mechanism-based enzyme inactivation: application to nitric oxide synthase. AAPS PharmSci 2:E8

    Article  CAS  PubMed  Google Scholar 

  29. Kitz R, Wilson IB (1962) Esters of methanesulfonic acid as irreversible inhibitors of acetylcholinesterase. J Biol Chem 237:3245–3249

    CAS  PubMed  Google Scholar 

  30. Gal S, Abassi ZA, Youdim MB (2010) Limited potentiation of blood pressure in response to oral tyramine by the Anti-Parkinson Brain Selective Multifunctional Monoamine Oxidase-AB Inhibitor, M30. Neurotox Res 18(2):143–150

    Article  CAS  PubMed  Google Scholar 

  31. Gal S, Zheng H, Fridkin M et al (2005) Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. In vivo selective brain monoamine oxidase inhibition and prevention of MPTP-induced striatal dopamine depletion. J Neurochem 95:79–88

    Article  CAS  PubMed  Google Scholar 

  32. Avramovich-Tirosh Y, Amit T, Bar-Am O et al (2007) Therapeutic targets and potential of the novel brain- permeable multifunctional iron chelator-monoamine oxidase inhibitor drug, M-30, for the treatment of Alzheimer’s disease. J Neurochem 100:490–502

    Article  CAS  PubMed  Google Scholar 

  33. Avramovich-Tirosh Y, Reznichenko L, Mit T et al (2007) Neurorescue activity, APP regulation and amyloid-beta peptide reduction by novel multi-functional brain permeable iron- chelating- antioxidants, M-30 and green tea polyphenol, EGCG. Curr Alzheimer Res 4:403–411

    Article  CAS  PubMed  Google Scholar 

  34. Bush AI (2008) Drug development based on the metals hypothesis of Alzheimer’s disease. J Alzheimers Dis 15:223–240

    CAS  PubMed  Google Scholar 

  35. Cavalli A, Bolognesi ML, Minarini A et al (2008) Multi-target-directed ligands to combat neurodegenerative diseases. J Med Chem 51:347–372

    Article  CAS  PubMed  Google Scholar 

  36. Schugar H, Green DE, Bowen ML et al (2007) Combating Alzheimer’s disease with multifunctional molecules designed for metal passivation. Angew Chem Int Ed Engl 46:1716–1718

    Article  CAS  PubMed  Google Scholar 

  37. Liu G, Men P, Kudo W et al (2009) Nanoparticle-chelator conjugates as inhibitors of amyloid-beta aggregation and neurotoxicity: a novel therapeutic approach for Alzheimer disease. Neurosci Lett 455:187–190

    Article  CAS  PubMed  Google Scholar 

  38. Charkoudian LK, Pham DM, Franz KJ (2006) A pro-chelator triggered by hydrogen peroxide inhibits iron-promoted hydroxyl radical formation. J Am Chem Soc 128:12424–12425

    Article  CAS  PubMed  Google Scholar 

  39. Folk DS, Franz KJ (2010) A prochelator activated by beta-secretase inhibits Abeta aggregation and suppresses copper-induced reactive oxygen species formation. J Am Chem Soc 132:4994–4995

    Article  CAS  PubMed  Google Scholar 

  40. Di Stefano A, Sozio P, Cocco A et al (2006) L-dopa- and dopamine-(R)-alpha-lipoic acid conjugates as multifunctional codrugs with antioxidant properties. J Med Chem 49:1486–1493

    Article  CAS  PubMed  Google Scholar 

  41. Rodriguez-Rodriguez C, Sanchez de Groot N, Rimola A et al (2009) Design, selection, and characterization of thioflavin-based intercalation compounds with metal chelating properties for application in Alzheimer’s disease. J Am Chem Soc 131:1436–1451

    Article  CAS  PubMed  Google Scholar 

  42. Hindo SS, Mancino AM, Braymer JJ et al (2009) Small molecule modulators of copper-induced Abeta aggregation. J Am Chem Soc 131:16663–16665

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We thank the Alzheimer Association (USA), Alzheimer Drug Discovery Foundation (New York, USA), Technion-Research and Development and the Weizmann Institute for generous support of this work.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Mati Fridkin or Moussa B. H. Youdim.

Additional information

Dedication to Abel Lathja

I do not recall exactly when I first met Abel, but I think it was in Budapest many years ego. What I recall was the fact he was so complimentary to my work as young neurochemist and his encouragement throughout the years I have known him. I was very much moved and honored when decided to dedicate one volume of Neurochemical Research for my contributions and he wrote the most flattering preface. Besides his tremendous scientific contributions, it is his Hungarian charm and gentleness that I so admire, including his unflenching friendship. This has made him many friends and admires from all the world. Moussa B.H. Youdim.

Special Issue: In Honor of Dr. Abel Lajtha.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Zheng, H., Fridkin, M. & Youdim, M.B.H. Site-Activated Chelators Derived from Anti-Parkinson Drug Rasagiline as a Potential Safer and More Effective Approach to the Treatment of Alzheimer’s Disease. Neurochem Res 35, 2117–2123 (2010). https://doi.org/10.1007/s11064-010-0293-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-010-0293-1

Keywords

Navigation