Skip to main content

Advertisement

Log in

Aroylhydrazones constitute a promising class of ‘metal-protein attenuating compounds’ for the treatment of Alzheimer’s disease: a proof-of-concept based on the study of the interactions between zinc(II) and pyridine-2-carboxaldehyde isonicotinoyl hydrazone

  • Original Paper
  • Published:
JBIC Journal of Biological Inorganic Chemistry Aims and scope Submit manuscript

Abstract

With the increasing life expectancy of the world’s population, neurodegenerative diseases, such as Alzheimer’s disease (AD), will become a much more relevant public health issue. This fact, coupled with the lack of efficacy of the available treatments, has been driving research directed to the development of new drugs for this pathology. Metal-protein attenuating compounds (MPACs) constitute a promising class of agents with potential application on the treatment of neurodegenerative diseases, such as AD. Currently, most MPACs are based on 8-hydroxyquinoline. Recently, our research group has described the hybrid aroylhydrazone containing the 8-hydroxyquinoline group INHHQ as a promising MPAC. By studying the known structure-related ligand HPCIH, which does not contain the phenol moiety, as a simplified chemical model for INHHQ, we aimed to clarify the real impact of the aroylhydrazone group for the MPAC activity of a compound with potential anti-Alzheimer’s activity. The present work describes a detailed solution and solid-state study of the coordination of HPCIH with Zn2+ ions, as well as its in vitro binding-ability towards this metal in the presence of the Aβ(1–40) peptide. Similar to INHHQ, HPCIH is able to efficiently compete with Aβ(1–40) for Zn2+ ions, performing as expected for an MPAC. The similarity between the behaviors of both ligands is remarkable. Taken together, the data presented herein point to aroylhydrazones, such as the compounds HPCIH and the previously published INHHQ, as encouraging MPACs for the treatment of AD.

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.

Institutional subscriptions

Scheme 1
Fig. 1
Fig. 2
Fig. 3
Scheme 2
Scheme 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

Abbreviations

Aβ:

β-Amyloid peptide

HPCIH:

Pyridine-2-carboxaldehyde isonicotinoyl hydrazone

INHHQ:

8-Hydroxyquinoline-2-carboxaldehyde isonicotinoyl hydrazone

MPACs:

Metal-protein attenuating compounds

References

  1. Hardy JA, Higgins GA (1992) Alzheimer’s disease: the amyloid cascade hypothesis. Science 256(5054):184–185

    Article  CAS  Google Scholar 

  2. Kayed R et al (2004) Permeabilization of lipid bilayers is a common conformation-dependent activity of soluble amyloid oligomers in protein misfolding diseases. J Biol Chem 279(45):46363–46366

    Article  CAS  Google Scholar 

  3. Klein WL, Krafft GA, Finch CE (2001) Targeting small Abeta oligomers: the solution to an Alzheimer’s disease conundrum? Trends Neurosci 24(4):219–224

    Article  CAS  Google Scholar 

  4. Lambert MP et al (1998) Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc Natl Acad Sci USA 95(11):6448–6453

    Article  CAS  Google Scholar 

  5. Hane F, Leonenko Z (2014) Effect of metals on kinetic pathways of amyloid-β aggregation. Biomolecules 4(1):101–116

    Article  Google Scholar 

  6. Hane F et al (2013) Cu(2+) affects amyloid-β (1–42) aggregation by increasing peptide-peptide binding forces. PLoS One 8(3):e59005

    Article  CAS  Google Scholar 

  7. Deibel MA, Ehmann WD, Markesbery WR (1996) Copper, iron, and zinc imbalances in severely degenerated brain regions in Alzheimer’s disease: possible relation to oxidative stress. J Neurol Sci 143(1–2):137–142

    Article  CAS  Google Scholar 

  8. Azimi S, Rauk A (2011) On the involvement of copper binding to the N terminus of the amyloid Beta Peptide of Alzheimer’s disease: a computational study on model systems. Int J Alzheimers Dis 2011:539762

    PubMed  PubMed Central  Google Scholar 

  9. Tõugu V, Karafin A, Palumaa P (2008) Binding of zinc(II) and copper(II) to the full-length Alzheimer’s amyloid-beta peptide. J Neurochem 104(5):1249–1259

    Article  Google Scholar 

  10. Huang X et al (2004) Trace metal contamination initiates the apparent auto-aggregation, amyloidosis, and oligomerization of Alzheimer’s Abeta peptides. J Biol Inorg Chem 9(8):954–960

    Article  CAS  Google Scholar 

  11. Craddock TJ et al (2012) The zinc dyshomeostasis hypothesis of Alzheimer’s disease. PLoS One 7(3):e33552

    Article  CAS  Google Scholar 

  12. Faller P, Hureau C (2009) Bioinorganic chemistry of copper and zinc ions coordinated to amyloid-beta peptide. Dalton Trans 7:1080–1094

    Article  Google Scholar 

  13. Miura T et al (2000) Metal binding modes of Alzheimer’s amyloid beta-peptide in insoluble aggregates and soluble complexes. Biochemistry 39(23):7024–7031

    Article  CAS  Google Scholar 

  14. Yang DS et al (2000) Examining the zinc binding site of the amyloid-beta peptide. Eur J Biochem 267(22):6692–6698

    Article  CAS  Google Scholar 

  15. Faller P, Hureau C (2012) A bioinorganic view of Alzheimer’s disease: when misplaced metal ions (re)direct the electrons to the wrong target. Chemistry 18(50):15910–15920

    Article  CAS  Google Scholar 

  16. Ventriglia M et al (2015) Zinc in Alzheimer’s disease: a meta-analysis of serum, plasma, and cerebrospinal fluid studies. J Alzheimers Dis 46(1):75–87

    Article  CAS  Google Scholar 

  17. Bucossi S et al (2011) Copper in Alzheimer’s disease: a meta-analysis of serum, plasma, and cerebrospinal fluid studies. J Alzheimers Dis 24(1):175–185

    Article  CAS  Google Scholar 

  18. Scott LE, Orvig C (2009) Medicinal inorganic chemistry approaches to passivation and removal of aberrant metal ions in disease. Chem Rev 109(10):4885–4910

    Article  CAS  Google Scholar 

  19. Gaeta A et al (2011) Synthesis, physical-chemical characterisation and biological evaluation of novel 2-amido-3-hydroxypyridin-4(1H)-ones: iron chelators with the potential for treating Alzheimer’s disease. Bioorg Med Chem 19(3):1285–1297

    Article  CAS  Google Scholar 

  20. Budimir A (2011) Metal ions, Alzheimer’s disease and chelation therapy. Acta Pharm 61(1):1–14

    Article  CAS  Google Scholar 

  21. Cherny RA et al (2001) Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer’s disease transgenic mice. Neuron 30(3):665–676

    Article  CAS  Google Scholar 

  22. Drew SC (2017) The case for abandoning therapeutic chelation of copper ions in Alzheimer’s disease. Front Neurosci 11:317

    Article  Google Scholar 

  23. Squitti R et al (2017) Commentary: the case for abandoning therapeutic chelation of copper ions in Alzheimer’s disease. Front Neurol 8:503

    Article  Google Scholar 

  24. de Freitas LV et al (2013) Structural and vibrational study of 8-hydroxyquinoline-2-carboxaldehyde isonicotinoyl hydrazone—a potential metal-protein attenuating compound (MPAC) for the treatment of Alzheimer’s disease. Spectrochim Acta A Mol Biomol Spectrosc 116:41–48

    Article  Google Scholar 

  25. Hauser-Davis RA et al (2015) Disruption of zinc and copper interactions with Aβ(1–40) by a non-toxic, isoniazid-derived, hydrazone: a novel biometal homeostasis restoring agent in Alzheimer’s disease therapy? Metallomics 7:743–747

    Article  CAS  Google Scholar 

  26. Cukierman DS et al (2017) A moderate metal-binding hydrazone meets the criteria for a bioinorganic approach towards Parkinson’s disease: therapeutic potential, blood-brain barrier crossing evaluation and preliminary toxicological studies. J Inorg Biochem 170:160–168

    Article  CAS  Google Scholar 

  27. Becker E, Richardson DR (1999) Development of novel aroylhydrazone ligands for iron chelation therapy: 2-pyridylcarboxaldehyde isonicotinoyl hydrazone analogs. J Lab Clin Med 134(5):510–521

    Article  CAS  Google Scholar 

  28. Armstrong Claire M et al (2003) Structural variations and formation constants of first-row transition metal complexes of biologically active aroylhydrazones. Eur J Inorg Chem 2003(6):1145–1156

    Article  Google Scholar 

  29. Mondal S, Naskar S, Deya AK, Sinn E, Eribalb C, Herronc SR, Chattopadhyaya SK (2013) Mononuclear and binuclear Cu(II) complexes of some tridentate aroyl hydrazones. X-ray crystal structures of a mononuclear and a binuclear complex. Inorg Chim Acta 398:98–105

    Article  CAS  Google Scholar 

  30. Khandar AA, Afkhami FA, Hosseini-Yazdi SA, White JM, Kassel S, Dougherty WG, Lipkowski J, Van Derveer D, Giester G, Costantino F (2015) Anion influence in the structural diversity of cadmium coordination polymers constructed from a pyridine based Schiff base ligand. Inorg Chim Acta 427:87–96

    Article  CAS  Google Scholar 

  31. Li L, Zhang YZ, Liu E, Yang C, Golen JA, Rheingold AL, Zhang G (2016) Synthesis and structural characterization of zinc(II) and cobalt(II) complexes based on multidentate hydrazone ligands. J Mol Struct 1110:180–184

    Article  CAS  Google Scholar 

  32. Regland B et al (2001) Treatment of Alzheimer’s disease with clioquinol. Dement Geriatr Cogn Disord 12(6):408–414

    Article  CAS  Google Scholar 

  33. Adlard PA 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(1):43–55

    Article  CAS  Google Scholar 

  34. Zheng H 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(1):68–78

    Article  CAS  Google Scholar 

  35. Gomes LM et al (2014) 8-Hydroxyquinoline Schiff-base compounds as antioxidants and modulators of copper-mediated Aβ peptide aggregation. J Inorg Biochem 139:106–116

    Article  CAS  Google Scholar 

  36. Richardson D, Bernhardt PV, Becker EM (2006) Iron chelators and uses thereof. US6989397 B1

  37. Bruker (2004) APEX2 user manual. Version 1.22 ed

  38. Bruker (1999) SAINT Integration Software. Bruker AXS Inc., Madison

    Google Scholar 

  39. Sheldrick GM (2015) Crystal structure refinement with SHELXL. Acta Crystallogr C Struct Chem 71(Pt 1):3–8

    Article  Google Scholar 

  40. Sheldrick GM (2008) A short history of SHELX. Acta Crystallogr A 64(Pt 1):112–122

    Article  CAS  Google Scholar 

  41. International Tables for Crystallography C (2006)

  42. Dolomanov et al (2009) OLEX2: a complete structure solution, refinement and analysis program. J Appl Crystallogr 42(2):339–341

    Article  CAS  Google Scholar 

  43. Nakacjawa I, Shimanouchi T (1964) Infrared absorption spectra of aquo complexes and the nature of co-ordination bands. Spectrochim Acta 20:429–439

    Article  Google Scholar 

  44. Bryson D, Nuttall RH (1970) The far infrared spectra of five co-ordinate complexes. Spectrochim Acta Part A 26:2275–2280

    Article  CAS  Google Scholar 

  45. Frank CW, Rogers LB (1966) Infrared Spectral study of metal-pyridine, -substituted pyridine, and -quinoline complexes in the 667–150 cm−1 Region. Inorg Chem 5:615–622

    Article  CAS  Google Scholar 

  46. Díaz-Torres R, Coordinating AS (2011) Ability of anions and solvents towards transition metals and lanthanides. Dalton Trans 40:10742–10750

    Article  Google Scholar 

  47. Bernhardt PV et al (2007) Hydrazone chelators for the treatment of iron overload disorders: iron coordination chemistry and biological activity. Dalton Trans 30:3232–3244

    Article  Google Scholar 

  48. Bush AI, Tanzi RE (2008) Therapeutics for Alzheimer’s disease based on the metal hypothesis. Neurotherapeutics 5(3):421–432

    Article  CAS  Google Scholar 

  49. Talmard C, Bouzan A, Faller P (2007) Zinc binding to amyloid-beta: isothermal titration calorimetry and Zn competition experiments with Zn sensors. Biochemistry 46(47):13658–13666

    Article  CAS  Google Scholar 

  50. Pajouhesh H, Lenz GR (2005) Medicinal chemical properties of successful central nervous system drugs. NeuroRx 2(4):541–553

    Article  Google Scholar 

Download references

Acknowledgements

Nicolás A. Rey wishes to thank FAPERJ (Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro, Brazil) and CNPq (Conselho Nacional de Desenvolvimento Científico e Tecnológico, Brazil) for the research fellowships awarded. Daphne S. Cukierman thanks CNPq and CAPES (Coordenação de Aperfeiçoamento de Pessoal de Nível Superior, Brazil) for the scholarships granted. The authors are indebted to the Central Analytical Facility of the Department of Chemistry (PUC-Rio). The polycrystal X-ray diffraction analyses were done with the help of Prof. Dr. Roberto Avillez, at the X-Ray Diffraction Laboratory of the Chemical and Materials Engineering Department of PUC-Rio. The authors also thank LAMEM-UFF (Laboratório Multiusuário de Espectrometria de Massas da Universidade Federal Fluminense) for the mass spectrometry analyses.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nicolás A. Rey.

Electronic supplementary material

Below is the link to the electronic supplementary material.

775_2018_1606_MOESM1_ESM.pdf

Crystallographic data for complex (2) has been deposited with the Cambridge Crystallographic Data Centre as Supplementary Publications CCDC No. 1849711, respectively. Copies of the data can be obtained free of charge on application to the CCDC (http://www.ccdc.cam.ac.uk/conts/retrieving.html). Thermogravimetric curve (Fig. S1) and UV–Vis stability (Fig. S13) of HPCIH, as well as infrared and Raman (Figs. S2 and S3), and uni- and bidimensional NMR (Figs. S4 to S12) spectra of both HPCIH and complex (1) can be found as supplementary materials (PDF 912 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cukierman, D.S., Accardo, E., Gomes, R.G. et al. Aroylhydrazones constitute a promising class of ‘metal-protein attenuating compounds’ for the treatment of Alzheimer’s disease: a proof-of-concept based on the study of the interactions between zinc(II) and pyridine-2-carboxaldehyde isonicotinoyl hydrazone. J Biol Inorg Chem 23, 1227–1241 (2018). https://doi.org/10.1007/s00775-018-1606-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00775-018-1606-0

Keywords

Navigation