1
|
Kryl'skii ED, Razuvaev GA, Popova TN, Oleinik SA, Medvedeva SM, Shikhaliev KS. 6-Hydroxy-2,2,4-trimethyl-1,2,3,4-tetrahydroquinoline Demonstrates Neuroprotective Properties in Experimental Parkinson's Disease by Enhancing the Antioxidant System, Normalising Chaperone Activity and Suppressing Apoptosis. Neurochem Res 2024; 49:1387-1405. [PMID: 38502411 DOI: 10.1007/s11064-024-04125-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 03/21/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, whereby disturbances within the antioxidant defence system, increased aggregation of proteins, and activation of neuronal apoptosis all have a crucial role in the pathogenesis. In this context, exploring the neuroprotective capabilities of compounds that sustain the effectiveness of cellular defence systems in neurodegenerative disorders is worthwhile. During this study, we assessed how 6-hydroxy-2,2,4-trimethyl-1,2,3,4-tetrahydroquinoline (HTHQ), which has antioxidant properties, affects the functioning of the antioxidant system, the activity of NADPH-generating enzymes and chaperones, and the level of apoptotic processes in rats with rotenone-induced PD. Six groups of animals were formed for our experiment, each with 12 animals. These were: a control group, animals with rotenone-induced PD, rats with PD given HTHQ at a dose of 50 mg/kg, rats with PD given HTHQ at a dose of 25 mg/kg, animals with pathology who were administered a comparison drug rasagiline, and control animals who were administered HTHQ at a dose of 50 mg/kg. The study results indicate that administering HTHQ led to a significant decrease in oxidative stress in PD rats. The enhanced redox status in animal tissues was linked with the recovery of antioxidant enzyme activities and NADPH-generating enzyme function, as well as an upsurge in the mRNA expression levels of antioxidant genes and factors Nrf2 and Foxo1. Administering HTHQ to rats with PD normalized the chaperone-like activity and mRNA levels of heat shock protein 70. Rats treated with the compound displayed lower apoptosis intensity when compared to animals with pathology. Therefore, owing to its antioxidant properties, HTHQ demonstrated a beneficial impact on the antioxidant system, resulting in decreased requirements for chaperone activation and the inhibition of apoptosis processes triggered in PD. HTHQ at a dose of 50 mg/kg had a greater impact on the majority of the examined variables compared to rasagiline.
Collapse
Affiliation(s)
- Evgenii D Kryl'skii
- Department of Medical Biochemistry, Molecular and Cell Biology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018.
| | - Grigorii A Razuvaev
- Department of Medical Biochemistry, Molecular and Cell Biology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| | - Tatyana N Popova
- Department of Medical Biochemistry, Molecular and Cell Biology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| | - Sergei A Oleinik
- Department of Medical Biochemistry, Molecular and Cell Biology, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| | - Svetlana M Medvedeva
- Department of Organic Chemistry, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| | - Khidmet S Shikhaliev
- Department of Organic Chemistry, Voronezh State University, Universitetskaya Sq. 1, Voronezh, Russia, 394018
| |
Collapse
|
2
|
Ansari MA, Tripathi T, Venkidasamy B, Monziani A, Rajakumar G, Alomary MN, Alyahya SA, Onimus O, D'souza N, Barkat MA, Al-Suhaimi EA, Samynathan R, Thiruvengadam M. Multifunctional Nanocarriers for Alzheimer's Disease: Befriending the Barriers. Mol Neurobiol 2024; 61:3042-3089. [PMID: 37966683 DOI: 10.1007/s12035-023-03730-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
Neurodegenerative diseases (NDDs) have been increasing in incidence in recent years and are now widespread worldwide. Neuronal death is defined as the progressive loss of neuronal structure or function which is closely associated with NDDs and represents the intrinsic features of such disorders. Amyotrophic lateral sclerosis, frontotemporal dementia, Alzheimer's, Parkinson's, and Huntington's diseases (AD, PD, and HD, respectively) are considered neurodegenerative diseases that affect a large number of people worldwide. Despite the testing of various drugs, there is currently no available therapy that can remedy or effectively slow the progression of these diseases. Nanomedicine has the potential to revolutionize drug delivery for the management of NDDs. The use of nanoparticles (NPs) has recently been developed to improve drug delivery efficiency and is currently subjected to extensive studies. Nanoengineered particles, known as nanodrugs, can cross the blood-brain barrier while also being less invasive compared to the most treatment strategies in use. Polymeric, magnetic, carbonic, and inorganic NPs are examples of NPs that have been developed to improve drug delivery efficiency. Primary research studies using NPs to cure AD are promising, but thorough research is needed to introduce these approaches to clinical use. In the present review, we discussed the role of metal-based NPs, polymeric nanogels, nanocarrier systems such as liposomes, solid lipid NPs, polymeric NPs, exosomes, quantum dots, dendrimers, polymersomes, carbon nanotubes, and nanofibers and surfactant-based systems for the therapy of neurodegenerative diseases. In addition, we highlighted nanoformulations such as N-butyl cyanoacrylate, poly(butyl cyanoacrylate), D-penicillamine, citrate-coated peptide, magnetic iron oxide, chitosan (CS), lipoprotein, ceria, silica, metallic nanoparticles, cholinesterase inhibitors, an acetylcholinesterase inhibitors, metal chelators, anti-amyloid, protein, and peptide-loaded NPs for the treatment of AD.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Takshashila Tripathi
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Alan Monziani
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Govindasamy Rajakumar
- Department of Orthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology, 11442, Riyadh, Saudi Arabia
| | - Sami A Alyahya
- Wellness and Preventive Medicine Institute, King Abdulaziz City for Science and Technology, 11442, Riyadh, Saudi Arabia
| | - Oriane Onimus
- Faculty of Basic and Biomedical Sciences, University of Paris, Paris, France
| | - Naomi D'souza
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin, Saudi Arabia
| | - Ebtesam A Al-Suhaimi
- Research Consultation Department, Vice Presidency for Scientific Research and Innovation, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Ramkumar Samynathan
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
3
|
Du B, Chen K, Wang W, Lei P. Targeting Metals in Alzheimer's Disease: An Update. J Alzheimers Dis 2024; 101:S141-S154. [PMID: 39422951 DOI: 10.3233/jad-240140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
One pathological feature of Alzheimer's disease (AD) is the dysregulated metal ions, e.g., zinc, copper, and iron in the affected brain regions. The dysregulation of metal homeostasis may cause neurotoxicity and directly addressing these dysregulated metals through metal chelation or mitigating the downstream neurotoxicity stands as a pivotal strategy for AD therapy. This review aims to provide an up-to-date comprehensive overview of the application of metal chelators and drugs targeting metal-related neurotoxicity, such as antioxidants (ferroptotic inhibitors), in the context of AD treatment. It encompasses an exploration of their pharmacological effects, clinical research progress, and potential underlying mechanisms.
Collapse
Affiliation(s)
- Bin Du
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Kang Chen
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Weiwei Wang
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Liu X, Duan S, Jin Y, Walker E, Tsao M, Jang JH, Chen Z, Singh AK, Cantrell KL, Ingolfsson HI, Buratto SK, Bowers MT. Computationally Designed Molecules Modulate ALS-Related Amyloidogenic TDP-43 307-319 Aggregation. ACS Chem Neurosci 2023; 14:4395-4408. [PMID: 38050862 DOI: 10.1021/acschemneuro.3c00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023] Open
Abstract
Abnormal cytosolic aggregation of TAR DNA-binding protein of 43 kDa (TDP-43) is observed in multiple diseases, including amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration, and Alzheimer's disease. Previous studies have shown that TDP-43307-319 located at the C-terminal of TDP-43 can form higher-order oligomers and fibrils. Of particular interest are the hexamers that adopt a cylindrin structure that has been strongly correlated to neurotoxicity. In this study, we use the joint pharmacophore space (JPS) model to identify and generate potential TDP-43 inhibitors. Five JPS-designed molecules are evaluated using both experimental and computational methods: ion mobility mass spectrometry, thioflavin T fluorescence assay, circular dichroism spectroscopy, atomic force microscopy, and molecular dynamics simulations. We found that all five molecules can prevent the amyloid fibril formation of TDP-43307-319, but their efficacy varies significantly. Furthermore, among the five molecules, [AC0101] is the most efficient in preventing the formation of higher-order oligomers and dissociating preformed higher-order oligomers. Molecular dynamics simulations show that [AC0101] both is the most flexible and forms the most hydrogen bonds with the TDP-43307-319 monomer. The JPS-designed molecules can insert themselves between the β-strands in the hexameric cylindrin structure of TDP-43307-319 and can open its structure. Possible mechanisms for JPS-designed molecules to inhibit and dissociate TDP-43307-319 oligomers on an atomistic scale are proposed.
Collapse
Affiliation(s)
- Xikun Liu
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Shuya Duan
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Yingying Jin
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Ethan Walker
- Department of Chemistry, Westmont College, Santa Barbara, California 93108, United States
| | - Michelle Tsao
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Joshua H Jang
- Department of Chemistry, Westmont College, Santa Barbara, California 93108, United States
| | - Ziying Chen
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Ambuj K Singh
- Department of Computer Science, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Kristi Lazar Cantrell
- Department of Chemistry, Westmont College, Santa Barbara, California 93108, United States
| | - Helgi I Ingolfsson
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California 94550, United States
| | - Steven K Buratto
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Michael T Bowers
- Department of Chemistry & Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
5
|
Jin Y, Downey MA, Singh A, Buratto SK, Bowers MT. Computationally Designed Small Molecules Disassemble Both Soluble Oligomers and Protofibrils of Amyloid β-Protein Responsible for Alzheimer's Disease. ACS Chem Neurosci 2023; 14:2717-2726. [PMID: 37442126 DOI: 10.1021/acschemneuro.3c00266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/15/2023] Open
Abstract
Alzheimer's disease (AD) is one of the world's most pressing health crises. AD is an incurable disease affecting more than 6.5 million Americans, predominantly the elderly, and in its later stages, leads to memory loss, dementia, and death. Amyloid β (Aβ) protein aggregates have been one of the pathological hallmarks of AD since its initial characterization. The early stages of Aβ accumulation and aggregation involve the formation of oligomers, which are considered neurotoxic and play a key role in further aggregation into fibrils that eventually appear in the brain as amyloid plaques. We have recently shown by combining ion mobility mass spectrometry (IM-MS) and atomic force microscopy (AFM) that Aβ42 rapidly forms dodecamers (12-mers) as the terminal oligomeric state, and these dodecamers seed the early formation of Aβ42 protofibrils. The link between soluble oligomers and fibril formation is one of the essential aspects for understanding the root cause of the disease state and is critical to developing therapeutic interventions. Utilizing a joint pharmacophore space (JPS) method, potential drugs have been designed specifically for amyloid-related diseases. These small molecules were generated based on crucial chemical features necessary for target selectivity. In this paper, we utilize our combined IM-MS and AFM methods to investigate the impact of three second-generation JPS small-molecule inhibitors, AC0201, AC0202, and AC0203, on dodecamer as well as fibril formation in Aβ42. Our results indicate that AC0201 works well as an inhibitor and remodeler of both dodecamers and fibril formation, AC0203 behaves less efficiently, and AC0202 is ineffective.
Collapse
Affiliation(s)
- Yingying Jin
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Matthew A Downey
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Ambuj Singh
- Department of Computer Science, University of California, Santa Barbara, California 93106, United States
- Acelot, Inc., 3160 Porter Drive, Suite 200, Palo Alto, California 94304, United States
| | - Steven K Buratto
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Michael T Bowers
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
6
|
Nasr T, Kariuki BM, Elansary MM, Elhaggar R, Zaghary W. Synthesis and crystal structures of ( E)- N'-(4-chloro-3-nitro-benzyl-idene)acetohydrazide and ( E)-2-(4-chloro-benzyl-idene)-1-(quinolin-8-yl)hydrazine. Acta Crystallogr E Crystallogr Commun 2023; 79:762-766. [PMID: 37601392 PMCID: PMC10439415 DOI: 10.1107/s2056989023006412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023]
Abstract
The syntheses of two benzyl-idenehydrazine derivatives, namely, (E)-N'-(4-chloro-3-nitro-benzyl-idene)acetohydrazide, C9H8ClN3O3, and (E)-2-(4-chloro-benzyl-idene)-1-(quinolin-8-yl)hydrazine, C16H12ClN3, are reported. The mol-ecules have been characterized using IR, 1H NMR, 13C NMR and mass spectro-scopic and elemental analysis techniques, and their structures have been determined by single-crystal X-ray diffraction.
Collapse
Affiliation(s)
- Tamer Nasr
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, 11795 Helwan, Cairo, Egypt
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information, MTI, Cairo, Egypt
| | - Benson M. Kariuki
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff, CF10 3AT, United Kingdom
| | - Mai M. Elansary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, 11795 Helwan, Cairo, Egypt
| | - Radwan Elhaggar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, 11795 Helwan, Cairo, Egypt
| | - Wafaa Zaghary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, 11795 Helwan, Cairo, Egypt
| |
Collapse
|
7
|
Summers KL, Roseman G, Schilling KM, Dolgova NV, Pushie MJ, Sokaras D, Kroll T, Harris HH, Millhauser GL, Pickering IJ, George GN. Alzheimer's Drug PBT2 Interacts with the Amyloid β 1-42 Peptide Differently than Other 8-Hydroxyquinoline Chelating Drugs. Inorg Chem 2022; 61:14626-14640. [PMID: 36073854 PMCID: PMC9957665 DOI: 10.1021/acs.inorgchem.2c01694] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although Alzheimer's disease (AD) was first described over a century ago, it remains the leading cause of age-related dementia. Innumerable changes have been linked to the pathology of AD; however, there remains much discord regarding which might be the initial cause of the disease. The "amyloid cascade hypothesis" proposes that the amyloid β (Aβ) peptide is central to disease pathology, which is supported by elevated Aβ levels in the brain before the development of symptoms and correlations of amyloid burden with cognitive impairment. The "metals hypothesis" proposes a role for metal ions such as iron, copper, and zinc in the pathology of AD, which is supported by the accumulation of these metals within amyloid plaques in the brain. Metals have been shown to induce aggregation of Aβ, and metal ion chelators have been shown to reverse this reaction in vitro. 8-Hydroxyquinoline-based chelators showed early promise as anti-Alzheimer's drugs. Both 5-chloro-7-iodo-8-hydroxyquinoline (CQ) and 5,7-dichloro-2-[(dimethylamino)methyl]-8-hydroxyquinoline (PBT2) underwent unsuccessful clinical trials for the treatment of AD. To gain insight into the mechanism of action of 8HQs, we have investigated the potential interaction of CQ, PBT2, and 5,7-dibromo-8-hydroxyquinoline (B2Q) with Cu(II)-bound Aβ(1-42) using X-ray absorption spectroscopy (XAS), high energy resolution fluorescence detected (HERFD) XAS, and electron paramagnetic resonance (EPR). By XAS, we found CQ and B2Q sequestered ∼83% of the Cu(II) from Aβ(1-42), whereas PBT2 sequestered only ∼59% of the Cu(II) from Aβ(1-42), suggesting that CQ and B2Q have a higher relative Cu(II) affinity than PBT2. From our EPR, it became clear that PBT2 sequestered Cu(II) from a heterogeneous mixture of Cu(II)Aβ(1-42) species in solution, leaving a single Cu(II)Aβ(1-42) species. It follows that the Cu(II) site in this Cu(II)Aβ(1-42) species is inaccessible to PBT2 and may be less solvent-exposed than in other Cu(II)Aβ(1-42) species. We found no evidence to suggest that these 8HQs form ternary complexes with Cu(II)Aβ(1-42).
Collapse
Affiliation(s)
- Kelly L. Summers
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham Roseman
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Kevin M. Schilling
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Natalia V. Dolgova
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
| | - M. Jake Pushie
- Department of Surgery, University of Saskatchewan, 103 Hospital Dr, Saskatoon, Saskatchewan S7N 0W8, Canada
| | - Dimosthenis Sokaras
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Thomas Kroll
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025, United States
| | - Hugh H. Harris
- Department of Chemistry, University of Adelaide, South Australia 5005, Australia
| | - Glenn L. Millhauser
- Department of Chemistry and Biochemistry, University of California, Santa Cruz, 1156 High Street, Santa Cruz, California 95064, United States
| | - Ingrid J. Pickering
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Graham N. George
- Molecular and Environmental Sciences Group, Department of Geological Sciences, College of Arts and Science, University of Saskatchewan, 114 Science Place, Saskatoon, Saskatchewan S7N 5E2, Canada
- Department of Chemistry, College of Arts and Science, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| |
Collapse
|
8
|
|
9
|
Limegrover CS, LeVine H, Izzo NJ, Yurko R, Mozzoni K, Rehak C, Sadlek K, Safferstein H, Catalano SM. Alzheimer's protection effect of A673T mutation may be driven by lower Aβ oligomer binding affinity. J Neurochem 2021; 157:1316-1330. [PMID: 33025581 PMCID: PMC8246829 DOI: 10.1111/jnc.15212] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022]
Abstract
Several mutations conferring protection against Alzheimer's disease (AD) have been described, none as profound as the A673T mutation, where carriers are four times less likely to get AD compared to noncarriers. This mutation results in reduced amyloid beta (Aβ) protein production in vitro and lower lifetime Aβ concentration in carriers. Better understanding of the protective mechanisms of the mutation may provide important insights into AD pathophysiology and identify productive therapeutic intervention strategies for disease modification. Aβ(1-42) protein forms oligomers that bind saturably to a single receptor site on neuronal synapses, initiating the downstream toxicities observed in AD. Decreased formation, toxicity, or stability of soluble Aβ oligomers, or reduction of synaptic binding of these oligomers, may combine with overall lower Aβ concentration to underlie A673T's disease protecting mechanism. To investigate these possibilities, we compared the formation rate of soluble oligomers made from Icelandic A673T mutant and wild type (wt) Aβ(1-42) synthetic protein, the amount and intensity of oligomer bound to mature primary rat hippocampal/cortical neuronal synapses, and the potency of bound oligomers to impact trafficking rate in neurons in vitro using a physiologically relevant oligomer preparation method. At equal protein concentrations, mutant protein forms approximately 50% or fewer oligomers of high molecular weight (>50 kDa) compared to wt protein. Mutant oligomers are twice as potent at altering the cellular vesicle trafficking rate as wt at equivalent concentrations, however, mutant oligomers have a >4-fold lower binding affinity to synaptic receptors (Kd = 1,950 vs. 442 nM). The net effect of these differences is a lower overall toxicity at a given concentration. This study demonstrates for the first time that mutant A673T Aβ oligomers prepared with this method have fundamentally different assembly characteristics and biological impact from wt protein and indicates that its disease protecting mechanism may result primarily from the mutant protein's much lower binding affinity to synaptic receptors. This suggests that therapeutics that effectively reduce oligomer binding to synapses in the brain may be beneficial in AD.
Collapse
Affiliation(s)
| | - Harry LeVine
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKYUSA
| | | | | | | | | | | | | | | |
Collapse
|
10
|
|
11
|
Zhang X, Zhong M, Zhao P, Zhang X, Li Y, Wang X, Sun J, Lan W, Sun H, Wang Z, Gao H. Screening a specific Zn(ii)-binding peptide for improving the cognitive decline of Alzheimer's disease in APP/PS1 transgenic mice by inhibiting Zn 2+-mediated amyloid protein aggregation and neurotoxicity. Biomater Sci 2020; 7:5197-5210. [PMID: 31588929 DOI: 10.1039/c9bm00676a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Zn2+ has been implicated in the progression of Alzheimer's disease (AD), as amyloid-β protein (Aβ) aggregation and neurotoxicity are mediated by zinc ions. Therefore, development of metal chelators for inhibiting and regulating metal-triggered Aβ aggregation has received attention as a strategy for treating AD. Here, we used an approach based on phage display to screen for a Zn(ii)-binding peptide that specifically blocks Zn-triggered Aβ aggregation. A fixed Zn(ii) resin was prepared using Ni-IDA affinity resin, and the target Zn(ii) was screened by interaction with a heptapeptide phage library. After negative biopanning against IDA and four rounds of positive biopanning against Zn(ii), high specificity Zn(ii)-binding phages were obtained. Through DNA sequencing and ELISA, 15 sets of Zn(ii)-binding peptides with high histidine contents were identified. We chose a highly specific peptide against Zn(ii) with the sequence of H-M-Q-T-N-H-H, and its abilities to chelate Zn2+ and inhibit Zn2+-mediated Aβ aggregation were assessed in vitro. We loaded the Zn(ii)-binding peptide onto PEG-modified chitosan nanoparticles (NPs) to improve the stability and the bioavailability of the Zn(ii) binding peptide. PEG-modified chitosan NPs loaded with Zn(ii)-binding peptide (PEG/PZn-CS NPs) reduced Zn2+ concentrations and Aβ secretion in mouse neuroblastoma (N)2a cells stably over-expressing the APP Swedish mutation (N2aswe). Zn2+-Induced neurotoxicity, oxidative stress, and apoptosis were attenuated by PEG/PZn-CS NPs. Intranasal administration of PEG/PZn-CS NPs improved the cognitive ability of APPswe/PS1d9 (APP/PS1) double-transgenic mice and reduced Aβ plaques in the mouse brain. This study indicated that a Zn(ii)-binding peptide and its NPs have promise as a potential anti-AD agent.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- College of Life and Health Sciences, Northeastern University, Shenyang, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Vinoth N, Kalaiarasi C, Kumaradhas P, Vadivel P, Lalitha A. Synthesis and Antibacterial Activity of New N‐Substituted Hexahydroquinolinone Derivatives and X‐Ray Crystallographic Studies. ChemistrySelect 2020. [DOI: 10.1002/slct.201904565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
| | | | | | - Pullar Vadivel
- Department of ChemistrySalem Sowdeswari College Salem- 636010 Tamilnadu India
| | - Appaswami Lalitha
- Department of ChemistryPeriyar University Salem- 636011 Tamilnadu India
| |
Collapse
|
13
|
Yu HJ, Zhao W, Zhou Y, Cheng GJ, Sun M, Wang L, Yu L, Liang SH, Ran C. Salen-based bifunctional chemosensor for copper (II) ions: Inhibition of copper-induced amyloid-β aggregation. Anal Chim Acta 2020; 1097:144-152. [DOI: 10.1016/j.aca.2019.10.072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 10/13/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023]
|
14
|
Ruotolo R, Minato I, La Vitola P, Artioli L, Curti C, Franceschi V, Brindani N, Amidani D, Colombo L, Salmona M, Forloni G, Donofrio G, Balducci C, Del Rio D, Ottonello S. Flavonoid-Derived Human Phenyl-γ-Valerolactone Metabolites Selectively Detoxify Amyloid-β Oligomers and Prevent Memory Impairment in a Mouse Model of Alzheimer's Disease. Mol Nutr Food Res 2020; 64:e1900890. [PMID: 31914208 DOI: 10.1002/mnfr.201900890] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/24/2019] [Indexed: 11/06/2022]
Abstract
SCOPE Amyloid-β oligomers (AβO) are causally related to Alzheimer's disease (AD). Dietary natural compounds, especially flavonoids and flavan-3-ols, hold great promise as potential AD-preventive agents but their host and gut microbiota metabolism complicates identification of the most relevant bioactive species. This study aims to investigate the ability of a comprehensive set of phenyl-γ-valerolactones (PVL), the main circulating metabolites of flavan-3-ols and related dietary compounds in humans, to prevent AβO-mediated toxicity. METHODS AND RESULTS The anti-AβO activity of PVLs is examined in different cell model systems using a highly toxic β-oligomer-forming polypeptide (β23) as target toxicant. Multiple PVLs, and particularly the monohydroxylated 5-(4'-hydroxyphenyl)-γ-valerolactone metabolite [(4'-OH)-PVL], relieve β-oligomer-induced cytotoxicity in yeast and mammalian cells. As revealed by atomic force microscopy (AFM) and other in vitro assays, (4'-OH)-PVL interferes with AβO (but not fibril) assembly and actively remodels preformed AβOs into nontoxic amorphous aggregates. In keeping with the latter mode of action, treatment of AβOs with (4'-OH)-PVL prior to brain injection strongly reduces memory deterioration as well as neuroinflammation in a mouse model of AβO-induced memory impairment. CONCLUSION PVLs, which have been validated as biomarkers of the dietary intake of flavan-3-ols, lend themselves as novel AβO-selective, candidate AD-preventing compounds.
Collapse
Affiliation(s)
- Roberta Ruotolo
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Ilaria Minato
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Pietro La Vitola
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Luisa Artioli
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Claudio Curti
- Department of Food and Drug, University of Parma, 43124, Parma, Italy
| | | | | | - Davide Amidani
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Gaetano Donofrio
- Department of Veterinary Science, University of Parma, 43126, Parma, Italy
| | - Claudia Balducci
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, 20156, Milan, Italy
| | - Daniele Del Rio
- Department of Veterinary Science, University of Parma, 43126, Parma, Italy
| | - Simone Ottonello
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124, Parma, Italy.,Biopharmanet-Tec, University of Parma, 43124, Parma, Italy
| |
Collapse
|
15
|
Laos V, Bishop D, Lang CA, Marsh NM, Cantrell KL, Buratto SK, Singh AK, Bowers MT. Modulating ALS-Related Amyloidogenic TDP-43 307-319 Oligomeric Aggregates with Computationally Derived Therapeutic Molecules. Biochemistry 2019; 59:499-508. [PMID: 31846303 DOI: 10.1021/acs.biochem.9b00905] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
TDP-43 aggregates are a salient feature of amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), and a variety of other neurodegenerative diseases, including Alzheimer's disease (AD). With an anticipated growth in the most susceptible demographic, projections predict neurodegenerative diseases will potentially affect 15 million people in the United States by 2050. Currently, there are no cures for ALS, FTD, or AD. Previous studies of the amyloidogenic core of TDP-43 have demonstrated that oligomers greater than a trimer are associated with toxicity. Utilizing a joint pharmacophore space (JPS) method, potential drugs have been designed specifically for amyloid-related diseases. These molecules were generated on the basis of key chemical features necessary for blood-brain barrier permeability, low adverse side effects, and target selectivity. Combining ion-mobility mass spectrometry and atomic force microscopy with the JPS computational method allows us to more efficiently evaluate a potential drug's efficacy in disrupting the development of putative toxic species. Our results demonstrate the dissociation of higher-order oligomers in the presence of these novel JPS-generated inhibitors into smaller oligomer species. Additionally, drugs approved by the Food and Drug Administration for the treatment of ALS were also evaluated and demonstrated to maintain higher-order oligomeric assemblies. Possible mechanisms for the observed action of the JPS molecules are discussed.
Collapse
Affiliation(s)
- Veronica Laos
- Department of Chemistry & Biochemistry , University of California, Santa Barbara , Santa Barbara , California 93106 , United States
| | - Dezmond Bishop
- Department of Chemistry & Biochemistry , University of California, Santa Barbara , Santa Barbara , California 93106 , United States
| | | | - Nicole M Marsh
- Department of Chemistry , Westmont College , Santa Barbaraa , California 93108 , United States
| | - Kristi Lazar Cantrell
- Department of Chemistry , Westmont College , Santa Barbaraa , California 93108 , United States
| | - Steven K Buratto
- Department of Chemistry & Biochemistry , University of California, Santa Barbara , Santa Barbara , California 93106 , United States
| | - Ambuj K Singh
- Department of Computer Science , University of California, Santa Barbara , Santa Barbara , California 93106-5110 , United States
| | - Michael T Bowers
- Department of Chemistry & Biochemistry , University of California, Santa Barbara , Santa Barbara , California 93106 , United States
| |
Collapse
|
16
|
Thamban Chandrika N, Fosso MY, Tsodikov OV, LeVine H, Garneau-Tsodikova S. Combining Chalcones with Donepezil to Inhibit Both Cholinesterases and Aβ Fibril Assembly. Molecules 2019; 25:E77. [PMID: 31878304 PMCID: PMC6983213 DOI: 10.3390/molecules25010077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/18/2019] [Accepted: 12/21/2019] [Indexed: 02/08/2023] Open
Abstract
The fact that the number of people with Alzheimer's disease is increasing, combined with the limited availability of drugs for its treatment, emphasize the need for the development of novel effective therapeutics for treating this brain disorder. Herein, we focus on generating 12 chalcone-donepezil hybrids, with the goal of simultaneously targeting amyloid-β (Aβ) peptides as well as cholinesterases (i.e., acetylcholinesterase (AChE) and butyrylcholinesterase (BChE)). We present the design, synthesis, and biochemical evaluation of these two series of novel 1,3-chalcone-donepezil (15a-15f) or 1,4-chalcone-donepezil (16a-16f) hybrids. We evaluate the relationship between their structures and their ability to inhibit AChE/BChE activity as well as their ability to bind Aβ peptides. We show that several of these novel chalcone-donepezil hybrids can successfully inhibit AChE/BChE as well as the assembly of N-biotinylated Aβ(1-42) oligomers. We also demonstrate that the Aβ binding site of these hybrids differs from that of Pittsburgh Compound B (PIB).
Collapse
Affiliation(s)
- Nishad Thamban Chandrika
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA; (N.T.C.); (M.Y.F.); (O.V.T.)
| | - Marina Y. Fosso
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA; (N.T.C.); (M.Y.F.); (O.V.T.)
| | - Oleg V. Tsodikov
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA; (N.T.C.); (M.Y.F.); (O.V.T.)
| | - Harry LeVine
- Center on Aging, School of Medicine, University of Kentucky, Lexington, KY 40536-0230, USA;
- Department of Molecular and Cellular Biochemistry, School of Medicine, University of Kentucky, Lexington, KY 40536-0230, USA
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0596, USA; (N.T.C.); (M.Y.F.); (O.V.T.)
| |
Collapse
|
17
|
Redox active metals in neurodegenerative diseases. J Biol Inorg Chem 2019; 24:1141-1157. [PMID: 31650248 DOI: 10.1007/s00775-019-01731-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022]
Abstract
Copper (Cu) and iron (Fe) are redox active metals essential for the regulation of cellular pathways that are fundamental for brain function, including neurotransmitter synthesis and release, neurotransmission, and protein turnover. Cu and Fe are tightly regulated by sophisticated homeostatic systems that tune the levels and localization of these redox active metals. The regulation of Cu and Fe necessitates their coordination to small organic molecules and metal chaperone proteins that restrict their reactions to specific protein centres, where Cu and Fe cycle between reduced (Fe2+, Cu+) and oxidised states (Fe3+, Cu2+). Perturbation of this regulation is evident in the brain affected by neurodegeneration. Here we review the evidence that links Cu and Fe dyshomeostasis to neurodegeneration as well as the promising preclinical and clinical studies reporting pharmacological intervention to remedy Cu and Fe abnormalities in the treatment of Alzheimer's disease (AD), Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS).
Collapse
|
18
|
Nam G, Lim MH. Intertwined Pathologies of Amyloid-β and Metal Ions in Alzheimer’s Disease: Metal–Amyloid-β. CHEM LETT 2019. [DOI: 10.1246/cl.190281] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Geewoo Nam
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- Department of Chemistry, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
19
|
Downey MA, Giammona MJ, Lang CA, Buratto SK, Singh A, Bowers MT. Inhibiting and Remodeling Toxic Amyloid-Beta Oligomer Formation Using a Computationally Designed Drug Molecule That Targets Alzheimer's Disease. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2019; 30:85-93. [PMID: 29713966 PMCID: PMC6258352 DOI: 10.1007/s13361-018-1975-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 05/25/2023]
Abstract
Alzheimer's disease (AD) is rapidly reaching epidemic status among a burgeoning aging population. Much evidence suggests the toxicity of this amyloid disease is most influenced by the formation of soluble oligomeric forms of amyloid β-protein, particularly the 42-residue alloform (Aβ42). Developing potential therapeutics in a directed, streamlined approach to treating this disease is necessary. Here we utilize the joint pharmacophore space (JPS) model to design a new molecule [AC0107] incorporating structural characteristics of known Aβ inhibitors, blood-brain barrier permeability, and limited toxicity. To test the molecule's efficacy experimentally, we employed ion mobility mass spectrometry (IM-MS) to discover [AC0107] inhibits the formation of the toxic Aβ42 dodecamer at both high (1:10) and equimolar concentrations of inhibitor. Atomic force microscopy (AFM) experiments reveal that [AC0107] prevents further aggregation of Aβ42, destabilizes preformed fibrils, and reverses Aβ42 aggregation. This trend continues for long-term interaction times of 2 days until only small aggregates remain with virtually no fibrils or higher order oligomers surviving. Pairing JPS with IM-MS and AFM presents a powerful and effective first step for AD drug development. Graphical Abstract.
Collapse
Affiliation(s)
- Matthew A Downey
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, 93106, USA
| | - Maxwell J Giammona
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, 93106, USA
| | - Christian A Lang
- Acelot, Inc., 5385 Hollister Ave, Suite 111, Santa Barbara, CA, 93111, USA
| | - Steven K Buratto
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, 93106, USA
| | - Ambuj Singh
- Acelot, Inc., 5385 Hollister Ave, Suite 111, Santa Barbara, CA, 93111, USA
- Department of Computer Science, University of California, Santa Barbara, CA, 93106, USA
| | - Michael T Bowers
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
20
|
Diagnostics and Treatments of Iron-Related CNS Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1173:179-194. [PMID: 31456211 DOI: 10.1007/978-981-13-9589-5_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Iron has been proposed to be responsible for neuronal loss in several diseases of the central nervous system, including Alzheimer's disease (AD), Parkinson's disease (PD), stroke, Friedreich's ataxia (FRDA), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS). In many diseases, abnormal accumulation of brain iron in disease-affected area has been observed, without clear knowledge of the contribution of iron overload to pathogenesis. Recent evidences implicate that key proteins involved in the disease pathogenesis may also participate in cellular iron metabolism, suggesting that the imbalance of brain iron homeostasis is associated with the diseases. Considering the complicated regulation of iron homeostasis within the brain, a thorough understanding of the molecular events leading to this phenotype is still to be investigated. However, current understanding has already provided the basis for the diagnosis and treatment of iron-related CNS diseases, which will be reviewed here.
Collapse
|
21
|
Villemagne VL, Rowe CC, Barnham KJ, Cherny R, Woodward M, Bozinosvski S, Salvado O, Bourgeat P, Perez K, Fowler C, Rembach A, Maruff P, Ritchie C, Tanzi R, Masters CL. A randomized, exploratory molecular imaging study targeting amyloid β with a novel 8-OH quinoline in Alzheimer's disease: The PBT2-204 IMAGINE study. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:622-635. [PMID: 29201996 PMCID: PMC5702880 DOI: 10.1016/j.trci.2017.10.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Introduction We are developing a second generation 8-OH quinoline (2-(dimethylamino) methyl-5, 7-dichloro-8-hydroxyquinoline [PBT2, Prana Biotechnology]) for targeting amyloid β (Aβ) in Alzheimer's disease (AD). In an earlier phase IIa, 3 month trial, PBT2 lowered cerebrospinal fluid Aβ by 13% and improved cognition (executive function) in a dose-related fashion in early AD. We, therefore, sought to learn whether PBT2 could alter the Aβ-PET signal in subjects with prodromal or mild AD, in an exploratory randomized study over a 12-month phase in a double-blind and a 12-month open label extension phase trial design. Methods For inclusion, the usual clinical criteria for prodromal or probable AD, Mini–Mental State Examination ≥20, and global Pittsburgh compound B (PiB)-PET standardized uptake volume ratio (SUVR) >1.7 were used. As this was an exploratory study, we included contemporaneous matched control data from the Australian Imaging Biomarker and Lifestyle Study (AIBL). Other measures included fluorodeoxyglucose-positron emission tomography, magnetic resonance imaging volumetrics, blood Aβ biomarkers, and cognition and function. Results Forty subjects completed the first 12-month double-blind phase (placebo = 15, PBT2 = 25), and 27 subjects completed the 12-month open label extension phase (placebo = 11, PBT2 = 16). Overall, PTB2 250 mg/day was safe and well tolerated. The mean PiB-PET SUVR at baseline was 2.51 ± 0.59. After adjusting for baseline SUVR, in the double-blind phase, the placebo group showed a nonsignificant decline in PiB-PET SUVR, whereas the PBT2 group declined significantly (P = .048). Subjects who did not enter or complete the extension study had a significantly higher 12-month Aβ-PET SUVR (2.68 ± 0.55) compared with those who completed (2.29 ± 0.48). Both groups differed significantly from the rate of change over 12 months in the AIBL control group. In the open label 12-month extension study, the PiB-SUVR stabilized. There were no significant differences between PBT2 and controls in fluorodeoxyglucose-positron emission tomography, magnetic resonance imaging volumetrics, blood Aβ biomarkers, or cognition/function over the course of the double-blind phase. Discussion There was no significant difference between PBT2 and controls at 12 months, likely due to the large individual variances over a relatively small number of subjects. PBT2 was associated with a significant 3% PiB-PET SUVR decline in the double-blind phase and a stabilization of SUVR in the open-label phase. From this exploratory study, we have learned that the entry criterion of SUVR should have been set at ≥ 1.5 and <2.0, where we know from the AIBL study that subjects in this band are accumulating Aβ in a linear fashion and that subjects who withdrew from this type of study have much higher SUVRs, which if not taken into account, could distort the final results. Because of large individual variations in SUVR, future studies of PBT2 will require larger numbers of subjects (n > 90 per arm) over a longer period (18 months or more). Further evaluation of higher doses of PBT2 in earlier stages of AD is warranted. Trial Registration ACTRN 12611001008910 and ACTRN 12613000777796. Exploratory randomized controlled trial for Alzheimer's disease using novel 8-OH quinoline. Use of Aβ positron emission tomography molecular imaging as intake criterion. Small numbers, large variances, and higher than expected mean baseline standardized uptake volume ratio may have contributed to lack of demonstrable efficacy.
Collapse
Affiliation(s)
- Victor L Villemagne
- Department of Molecular Imaging and Therapy (Research), Austin Health, Melbourne, Victoria, Australia.,The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christopher C Rowe
- Department of Molecular Imaging and Therapy (Research), Austin Health, Melbourne, Victoria, Australia.,The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kevin J Barnham
- The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Robert Cherny
- The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia.,Prana Biotechnology, Melbourne, Victoria, Australia
| | - Michael Woodward
- Medical and Cognitive Research Unit, Aged Care and Residential Services, Austin Health, Melbourne, Victoria, Australia
| | - Svetlana Bozinosvski
- Department of Molecular Imaging and Therapy (Research), Austin Health, Melbourne, Victoria, Australia
| | - Olivier Salvado
- Health and Biosecurity, CSIRO, Brisbane, Queensland, Australia
| | | | - Keyla Perez
- The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christopher Fowler
- The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alan Rembach
- The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia.,Cogstate Ltd, Melbourne, Victoria, Australia
| | - Craig Ritchie
- Psychiatry, University of Edinburgh, Edinburgh, Scotland, UK
| | - Rudy Tanzi
- Genetics and Aging Research Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
22
|
Horton W, Sood A, Peerannawar S, Kugyela N, Kulkarni A, Tulsan R, Tran CD, Soule J, LeVine H, Török B, Török M. Synthesis and application of β-carbolines as novel multi-functional anti-Alzheimer's disease agents. Bioorg Med Chem Lett 2017; 27:232-236. [PMID: 27923619 PMCID: PMC5282889 DOI: 10.1016/j.bmcl.2016.11.067] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 12/22/2022]
Abstract
The design, synthesis and assessment of β-carboline core-based compounds as potential multifunctional agents against several processes that are believed to play a significant role in Alzheimer's disease (AD) pathology, are described. The activity of the compounds was determined in Aβ self-assembly (fibril and oligomer formation) and cholinesterase (AChE, BuChE) activity inhibition, and their antioxidant properties were also assessed. To obtain insight into the mode of action of the compounds, HR-MS studies were carried out on the inhibitor-Aβ complex formation and molecular docking was performed on inhibitor-BuChE interactions. While several compounds exhibited strong activities in individual assays, compound 14 emerged as a promising multi-target lead for the further structure-activity relationship studies.
Collapse
Affiliation(s)
- William Horton
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA
| | - Abha Sood
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA
| | | | - Nandor Kugyela
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA
| | - Aditya Kulkarni
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA
| | - Rekha Tulsan
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA
| | - Chris D Tran
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA
| | - Jessica Soule
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA
| | - Harry LeVine
- Department of Molecular and Cellular Biochemistry, Chandler School of Medicine, and Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Béla Török
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA
| | - Marianna Török
- University of Massachusetts Boston, 100 Morrissey Blvd., Boston, MA, USA.
| |
Collapse
|
23
|
Proteins behaving badly. Substoichiometric molecular control and amplification of the initiation and nature of amyloid fibril formation: lessons from and for blood clotting. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017; 123:16-41. [DOI: 10.1016/j.pbiomolbio.2016.08.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/14/2016] [Accepted: 08/19/2016] [Indexed: 02/08/2023]
|
24
|
Wallin C, Luo J, Jarvet J, Wärmländer SKTS, Gräslund A. The Amyloid-β Peptide in Amyloid Formation Processes: Interactions with Blood Proteins and Naturally Occurring Metal Ions. Isr J Chem 2016. [DOI: 10.1002/ijch.201600105] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Cecilia Wallin
- Department of Biochemistry and Biophysics; Arrhenius Laboratories; Stockholm University; 10691 Stockholm Sweden
| | - Jinghui Luo
- Department of Biochemistry and Biophysics; Arrhenius Laboratories; Stockholm University; 10691 Stockholm Sweden
- Chemical Research Laboratory; University of Oxford; 12 Mansfield Road Oxford Ox 1 3TA UK
| | - Jüri Jarvet
- Department of Biochemistry and Biophysics; Arrhenius Laboratories; Stockholm University; 10691 Stockholm Sweden
- The National Institute of Chemical Physics and Biophysics; Tallinn Estonia
| | | | - Astrid Gräslund
- Department of Biochemistry and Biophysics; Arrhenius Laboratories; Stockholm University; 10691 Stockholm Sweden
| |
Collapse
|
25
|
8-Hydroxyquinolines in medicinal chemistry: A structural perspective. Eur J Med Chem 2016; 120:252-74. [DOI: 10.1016/j.ejmech.2016.05.007] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 01/12/2023]
|
26
|
Suwanjang W, Prachayasittikul S, Prachayasittikul V. Effect of 8-hydroxyquinoline and derivatives on human neuroblastoma SH-SY5Y cells under high glucose. PeerJ 2016; 4:e2389. [PMID: 27635352 PMCID: PMC5012261 DOI: 10.7717/peerj.2389] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/29/2016] [Indexed: 12/21/2022] Open
Abstract
8-Hydroxyquinoline and derivatives exhibit multifunctional properties, including antioxidant, antineurodegenerative, anticancer, anti-inflammatory and antidiabetic activities. In biological systems, elevation of intracellular calcium can cause calpain activation, leading to cell death. Here, the effect of 8-hydroxyquinoline and derivatives (5-chloro-7-iodo-8-hydroxyquinoline or clioquinol and 8-hydroxy-5-nitroquinoline or nitroxoline) on calpain-dependent (calpain-calpastatin) pathways in human neuroblastoma (SH-SY5Y) cells was investigated. 8-Hydroxyquinoline and derivatives ameliorated high glucose toxicity in SH-SY5Y cells. The investigated compounds, particularly clioquinol, attenuated the increased expression of calpain, even under high-glucose conditions. 8-Hydroxyquinoline and derivatives thus adversely affected the promotion of neuronal cell death by high glucose via the calpain-calpastatin signaling pathways. These findings support the beneficial effects of 8-hydroxyquinolines for further therapeutic development.
Collapse
Affiliation(s)
- Wilasinee Suwanjang
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University , Bangkok , Thailand
| | - Supaluk Prachayasittikul
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University , Bangkok , Thailand
| | - Virapong Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University , Bangkok , Thailand
| |
Collapse
|
27
|
Prati F, Bergamini C, Fato R, Soukup O, Korabecny J, Andrisano V, Bartolini M, Bolognesi ML. Novel 8-Hydroxyquinoline Derivatives as Multitarget Compounds for the Treatment of Alzheimer′s Disease. ChemMedChem 2016; 11:1284-95. [DOI: 10.1002/cmdc.201600014] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Federica Prati
- Department of Pharmacy and Biotechnology; Alma Mater Studiorum University of Bologna; Via Belmeloro 6/Via Irnerio 48 40126 Bologna Italy
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology; Alma Mater Studiorum University of Bologna; Via Belmeloro 6/Via Irnerio 48 40126 Bologna Italy
| | - Romana Fato
- Department of Pharmacy and Biotechnology; Alma Mater Studiorum University of Bologna; Via Belmeloro 6/Via Irnerio 48 40126 Bologna Italy
| | - Ondrej Soukup
- Biomedical Research Center; University Hospital Hradec Kralove; Sokolska 581 500 05 Hradec Kralove Czech Republic
| | - Jan Korabecny
- Biomedical Research Center; University Hospital Hradec Kralove; Sokolska 581 500 05 Hradec Kralove Czech Republic
| | - Vincenza Andrisano
- Department for Quality Life Studies; Alma Mater Studiorum University of Bologna; Corso d'Augusto 237 47921 Rimini Italy
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology; Alma Mater Studiorum University of Bologna; Via Belmeloro 6/Via Irnerio 48 40126 Bologna Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology; Alma Mater Studiorum University of Bologna; Via Belmeloro 6/Via Irnerio 48 40126 Bologna Italy
| |
Collapse
|
28
|
Aoki S, Ariyasu S, Hanaya K, Hisamatsu Y, Sugai T. Chemical Reactions of 8-Quinolinol Derivatives and Their Applications to Biochemical Tools and Enzyme Inhibitors. J SYN ORG CHEM JPN 2016. [DOI: 10.5059/yukigoseikyokaishi.74.482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Shin Aoki
- Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | - Shinya Ariyasu
- School of Physical & Mathematical Sciences, Nanyang Technological University
| | | | | | | |
Collapse
|
29
|
Hevroni BL, Major DT, Dixit M, Mhashal AR, Das S, Fischer B. Nucleoside-2′,3′/3′,5′-bis(thio)phosphate antioxidants are also capable of disassembly of amyloid beta42-Zn(ii)/Cu(ii) aggregates via Zn(ii)/Cu(ii)-chelation. Org Biomol Chem 2016; 14:4640-53. [DOI: 10.1039/c6ob00613b] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Nucleoside-2′,3′/3′,5′-bis(thio)phosphate antioxidants were identified as efficient agents of disassembly of Aβ42-Zn(ii)/Cu(ii) aggregates by M(ii)-chelation, thus making promising scaffolds for new Alzheimer's disease therapeutics.
Collapse
Affiliation(s)
| | - Dan Thomas Major
- Lise Meitner-Minerva Center of Computational Quantum Chemistry
- Bar-Ilan University
- Ramat-Gan 52900
- Israel
| | - Mudit Dixit
- Lise Meitner-Minerva Center of Computational Quantum Chemistry
- Bar-Ilan University
- Ramat-Gan 52900
- Israel
| | - Anil Ranu Mhashal
- Lise Meitner-Minerva Center of Computational Quantum Chemistry
- Bar-Ilan University
- Ramat-Gan 52900
- Israel
| | - Susanta Das
- Lise Meitner-Minerva Center of Computational Quantum Chemistry
- Bar-Ilan University
- Ramat-Gan 52900
- Israel
| | - Bilha Fischer
- Department of Chemistry
- Bar-Ilan University
- Ramat-Gan 52900
- Israel
| |
Collapse
|
30
|
Oliveri V, Bellia F, Grasso GI, Pietropaolo A, Vecchio G. Trehalose-8-hydroxyquinoline conjugates as antioxidant modulators of Aβ aggregation. RSC Adv 2016. [DOI: 10.1039/c6ra04204j] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The conjugation of trehalose with 8-hydroxyquinoline induces synergistic effects that lead to good antiaggregant ability. The difunctionalization of trehalose produces a better-performing antiaggregant compound.
Collapse
Affiliation(s)
- Valentina Oliveri
- Dipartimento di Scienze Chimiche
- Università di Catania
- Catania
- Italy
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici C.I.R.C.M.S.B
| | | | | | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute
- Università di Catanzaro
- 88100 Catanzaro
- Italy
| | | |
Collapse
|
31
|
MacLean MA, Diez-Cecilia E, Lavery CB, Reed MA, Wang Y, Weaver DF, Stradiotto M. Diversification of edaravone via palladium-catalyzed hydrazine cross-coupling: Applications against protein misfolding and oligomerization of beta-amyloid. Bioorg Med Chem Lett 2015; 26:100-4. [PMID: 26598460 DOI: 10.1016/j.bmcl.2015.11.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/05/2015] [Accepted: 11/08/2015] [Indexed: 11/15/2022]
Abstract
N-Aryl derivatives of edaravone were identified as potentially effective small molecule inhibitors of tau and beta-amyloid aggregation in the context of developing disease-modifying therapeutics for Alzheimer's disease (AD). Palladium-catalyzed hydrazine monoarylation protocols were then employed as an expedient means of preparing a focused library of 21 edaravone derivatives featuring varied N-aryl substitution, thereby enabling structure-activity relationship (SAR) studies. On the basis of data obtained from two functional biochemical assays examining the effect of edaravone derivatives on both fibril and oligomer formation, it was determined that derivatives featuring an N-biaryl motif were four-fold more potent than edaravone.
Collapse
Affiliation(s)
- Mark A MacLean
- Department of Chemistry, Dalhousie University, 6274 Coburg Road, PO Box 15000, Halifax, NS B3H 4R2, Canada
| | - Elena Diez-Cecilia
- Toronto Western Research Institute, University Health Network, University of Toronto, Krembil Discovery Tower, 4th Floor 4KD-472, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Christopher B Lavery
- Department of Chemistry, Dalhousie University, 6274 Coburg Road, PO Box 15000, Halifax, NS B3H 4R2, Canada
| | - Mark A Reed
- Treventis Corporation, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada
| | - Yanfei Wang
- Toronto Western Research Institute, University Health Network, University of Toronto, Krembil Discovery Tower, 4th Floor 4KD-472, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada
| | - Donald F Weaver
- Department of Chemistry, Dalhousie University, 6274 Coburg Road, PO Box 15000, Halifax, NS B3H 4R2, Canada; Toronto Western Research Institute, University Health Network, University of Toronto, Krembil Discovery Tower, 4th Floor 4KD-472, 60 Leonard Avenue, Toronto, ON M5T 2S8, Canada; Treventis Corporation, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada.
| | - Mark Stradiotto
- Department of Chemistry, Dalhousie University, 6274 Coburg Road, PO Box 15000, Halifax, NS B3H 4R2, Canada.
| |
Collapse
|
32
|
Potočňák I, Vranec P, Farkasová V, Sabolová D, Vataščinová M, Kudláčová J, Radojević ID, Čomić LR, Markovic BS, Volarevic V, Arsenijevic N, Trifunović SR. Low-dimensional compounds containing bioactive ligands. Part VI: Synthesis, structures, in vitro DNA binding, antimicrobial and anticancer properties of first row transition metal complexes with 5-chloro-quinolin-8-ol. J Inorg Biochem 2015; 154:67-77. [PMID: 26600190 DOI: 10.1016/j.jinorgbio.2015.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/12/2015] [Accepted: 10/28/2015] [Indexed: 01/19/2023]
Abstract
A series of new 3d metal complexes with 5-chloro-quinolin-8-ol (ClQ), [Mn(ClQ)2] (1), [Fe(ClQ)3] (2), [Co(ClQ)2(H2O)2] (3), [Ni(ClQ)2(H2O)2] (4), [Cu(ClQ)2] (5), [Zn(ClQ)2(H2O)2] (6), [Mn(ClQ)3]·DMF (7) and [Co(ClQ)3]·DMF·(EtOH)0.35 (8) (DMF=N,N-dimethylformamide), has been synthesized and characterized by elemental analysis, IR spectroscopy and TG-DTA thermal analysis. X-ray structure analysis of 7 and 8 revealed that these molecular complexes contain three chelate ClQ molecules coordinated to the central atoms in a deformed octahedral geometry and free space between the complex units is filled by solvated DMF and ethanol molecules. Antimicrobial activity of 1-6 was tested by determining the minimum inhibitory concentration and minimum microbicidal concentration against 12 strains of bacteria and 5 strains of fungi. The intensity of antimicrobial action varies depending on the group of microorganism and can be sorted: 1>ClQ>6>3/4>2>5. Complexes 1-6 exhibit high cytotoxic activity against MDA-MB, HCT-116 and A549 cancer cell lines. Among them, complex 2 is significantly more cytotoxic against MDA-MB cells than cisplatin at all tested concentrations and is not cytotoxic against control mesenchymal stem cells indicating that this complex seems to be a good candidate for future pharmacological evaluation. Interaction of 1-6 with DNA was investigated using UV-VIS spectroscopy, fluorescence spectroscopy and agarose gel electrophoresis. The binding studies indicate that 1-6 can interact with CT-DNA through intercalation; complex 2 has the highest binding affinity. Moreover, complexes 1-6 inhibit the catalytic activity of topoisomerase I.
Collapse
Affiliation(s)
- Ivan Potočňák
- Department of Inorganic Chemistry, Institute of Chemistry, P. J. Šafárik University, Moyzesova 11, SK-04154 Košice, Slovak Republic.
| | - Peter Vranec
- Department of Inorganic Chemistry, Institute of Chemistry, P. J. Šafárik University, Moyzesova 11, SK-04154 Košice, Slovak Republic
| | - Veronika Farkasová
- Department of Inorganic Chemistry, Institute of Chemistry, P. J. Šafárik University, Moyzesova 11, SK-04154 Košice, Slovak Republic
| | - Danica Sabolová
- Department of Biochemistry, Institute of Chemistry, P. J. Šafárik University, Moyzesova 11, SK-04154 Košice, Slovak Republic
| | - Michaela Vataščinová
- Department of Biochemistry, Institute of Chemistry, P. J. Šafárik University, Moyzesova 11, SK-04154 Košice, Slovak Republic
| | - Júlia Kudláčová
- Department of Biochemistry, Institute of Chemistry, P. J. Šafárik University, Moyzesova 11, SK-04154 Košice, Slovak Republic
| | - Ivana D Radojević
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Ljiljana R Čomić
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Bojana Simovic Markovic
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica, 34000 Kragujevac, Serbia
| | - Vladislav Volarevic
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica, 34000 Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica, 34000 Kragujevac, Serbia
| | - Srećko R Trifunović
- Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| |
Collapse
|
33
|
Aβ42-oligomer Interacting Peptide (AIP) neutralizes toxic amyloid-β42 species and protects synaptic structure and function. Sci Rep 2015; 5:15410. [PMID: 26510576 PMCID: PMC4625140 DOI: 10.1038/srep15410] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 09/22/2015] [Indexed: 12/21/2022] Open
Abstract
The amyloid-β42 (Aβ42) peptide is believed to be the main culprit in the pathogenesis of Alzheimer disease (AD), impairing synaptic function and initiating neuronal degeneration. Soluble Aβ42 oligomers are highly toxic and contribute to progressive neuronal dysfunction, loss of synaptic spine density, and affect long-term potentiation (LTP). We have characterized a short, L-amino acid Aβ-oligomer Interacting Peptide (AIP) that targets a relatively well-defined population of low-n Aβ42 oligomers, rather than simply inhibiting the aggregation of Aβ monomers into oligomers. Our data show that AIP diminishes the loss of Aβ42-induced synaptic spine density and rescues LTP in organotypic hippocampal slice cultures. Notably, the AIP enantiomer (comprised of D-amino acids) attenuated the rough-eye phenotype in a transgenic Aβ42 fly model and significantly improved the function of photoreceptors of these flies in electroretinography tests. Overall, our results indicate that specifically “trapping” low-n oligomers provides a novel strategy for toxic Aβ42-oligomer recognition and removal.
Collapse
|
34
|
Rational approach to an antiprion compound with a multiple mechanism of action. Future Med Chem 2015; 7:2113-20. [DOI: 10.4155/fmc.15.79] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: The main pathogenic event of prion disorders has been identified in the deposition of the disease-associated prion protein (PrPSc), which is accompanied by metal dyshomeostasis. Results: The multitarget-directed ligand 1, designed by combining a heteroaromatic prion recognition motif to an 8-hydroxyquinoline metal chelator, has been developed as a potential antiprion disease-modifying agent. Importantly, 1 was found to effectively clear PrPSc from scrapie-infected cells, and, at the same time, inhibit metal-induced prion aggregation and reactive oxygen species generation. 1 was also characterized in terms of pharmacokinetic properties in a preliminary in vitro investigation. Conclusion: Compound 1 has emerged as a suitable lead candidate against prion diseases and as a good starting point for a further optimization process.
Collapse
|
35
|
Oliveri V, Bellia F, Pietropaolo A, Vecchio G. Unusual Cyclodextrin Derivatives as a New Avenue to Modulate Self- and Metal-Induced Aβ Aggregation. Chemistry 2015; 21:14047-59. [PMID: 26298549 DOI: 10.1002/chem.201502155] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Indexed: 12/19/2022]
Abstract
Mounting evidence suggests an important role of cyclodextrins in providing protection in neurodegenerative disorders. Metal dyshomeostasis is reported to be a pathogenic factor in neurodegeneration because it could be responsible for damage involving oxidative stress and protein aggregation. As such, metal ions represent an effective target. To improve the metal-binding ability of cyclodextrin, we synthesized three new 8-hydroxyquinoline-cyclodextrin conjugates with difunctionalized cyclodextrins. In particular, the 3-difunctionalized regioisomer represents the first example of cyclodextrin with two pendants at the secondary rim, resulting in a promising compound. The derivatives have significant antioxidant capacity and the powerful activity in inhibiting self-induced amyloid-β aggregation seems to be led by synergistic effects of both cyclodextrin and hydroxyquinoline. Moreover, the derivatives are also able to complex metal ions and to inhibit metal-induced protein aggregation. Therefore, these compounds could have potential as therapeutic agents in diseases related to protein aggregation and metal dyshomeostasis.
Collapse
Affiliation(s)
- Valentina Oliveri
- Dipartimento di Scienze Chimiche, Università di Catania, Viale A. Doria 6, 95125, Catania (Italy).,Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici, C.I.R.C.M.S.B, Unità di Ricerca di Catania, 95125 Catania (Italy)
| | - Francesco Bellia
- Istituto di Biostrutture e Bioimmagini, CNR, Via P. Gaifami 18, 95126 Catania, Italy
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute, Università di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università di Catania, Viale A. Doria 6, 95125, Catania (Italy).
| |
Collapse
|
36
|
Fosso MY, LeVine H, Green KD, Tsodikov OV, Garneau-Tsodikova S. Effects of structural modifications on the metal binding, anti-amyloid activity, and cholinesterase inhibitory activity of chalcones. Org Biomol Chem 2015; 13:9418-26. [PMID: 26248214 DOI: 10.1039/c5ob01478f] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
As the number of individuals affected with Alzheimer's disease (AD) increases and the availability of drugs for AD treatment remains limited, the need to develop effective therapeutics for AD becomes more and more pressing. Strategies currently pursued include inhibiting acetylcholinesterase (AChE) and targeting amyloid-β (Aβ) peptides and metal-Aβ complexes. This work presents the design, synthesis, and biochemical evaluation of a series of chalcones, and assesses the relationship between their structures and their ability to bind metal ions and/or Aβ species, and inhibit AChE/BChE activity. Several chalcones were found to exhibit potent disaggregation of pre-formed N-biotinyl Aβ1-42 (bioAβ42) aggregates in vitro in the absence and presence of Cu(2+)/Zn(2+), while others were effective at inhibiting the action of AChE.
Collapse
Affiliation(s)
- Marina Y Fosso
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536-0596, USA.
| | | | | | | | | |
Collapse
|
37
|
Stabilization of nontoxic Aβ-oligomers: insights into the mechanism of action of hydroxyquinolines in Alzheimer's disease. J Neurosci 2015; 35:2871-84. [PMID: 25698727 DOI: 10.1523/jneurosci.2912-14.2015] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The extracellular accumulation of amyloid β (Aβ) peptides is characteristic of Alzheimer's disease (AD). However, formation of diffusible, oligomeric forms of Aβ, both on and off pathways to amyloid fibrils, is thought to include neurotoxic species responsible for synaptic loss and neurodegeneration, rather than polymeric amyloid aggregates. The 8-hydroxyquinolines (8-HQ) clioquinol (CQ) and PBT2 were developed for their ability to inhibit metal-mediated generation of reactive oxygen species from Aβ:Cu complexes and have both undergone preclinical and Phase II clinical development for the treatment of AD. Their respective modes of action are not fully understood and may include both inhibition of Aβ fibrillar polymerization and direct depolymerization of existing Aβ fibrils. In the present study, we find that CQ and PBT2 can interact directly with Aβ and affect its propensity to aggregate. Using a combination of biophysical techniques, we demonstrate that, in the presence of these 8-HQs and in the absence of metal ions, Aβ associates with two 8-HQ molecules and forms a dimer. Furthermore, 8-HQ bind Aβ with an affinity of 1-10 μm and suppress the formation of large (>30 kDa) oligomers. The stabilized low molecular weight species are nontoxic. Treatment with 8-HQs also reduces the levels of in vivo soluble oligomers in a Caenorhabditis elegans model of Aβ toxicity. We propose that 8-HQs possess an additional mechanism of action that neutralizes neurotoxic Aβ oligomer formation through stabilization of small (dimeric) nontoxic Aβ conformers.
Collapse
|
38
|
Bag S, Tulsan R, Sood A, Cho H, Redjeb H, Zhou W, LeVine H, Török B, Török M. Sulfonamides as multifunctional agents for Alzheimer’s disease. Bioorg Med Chem Lett 2015; 25:626-30. [DOI: 10.1016/j.bmcl.2014.12.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/03/2014] [Indexed: 11/30/2022]
|
39
|
Abstract
No disease modifying therapy exists for Alzheimer's disease (AD). The growing burden of this disease to our society necessitates continued investment in drug development. Over the last decade, multiple phase 3 clinical trials testing drugs that were designed to target established disease mechanisms of AD have all failed to benefit patients. There is, therefore, a need for new treatment strategies. Changes to the transition metals, zinc, copper, and iron, in AD impact on the molecular mechanisms of disease, and targeting these metals might be an alternative approach to treat the disease. Here we review how metals feature in molecular mechanisms of AD, and we describe preclinical and clinical data that demonstrate the potential for metal-based drug therapy.
Collapse
Affiliation(s)
- Scott Ayton
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, 3052 VIC Australia
| | - Peng Lei
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, 3052 VIC Australia
| | - Ashley I. Bush
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, 3052 VIC Australia
| |
Collapse
|
40
|
Zhang X, Tian Y, Li Z, Tian X, Sun H, Liu H, Moore A, Ran C. Design and synthesis of curcumin analogues for in vivo fluorescence imaging and inhibiting copper-induced cross-linking of amyloid beta species in Alzheimer's disease. J Am Chem Soc 2013; 135:16397-409. [PMID: 24116384 DOI: 10.1021/ja405239v] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this article, we first designed and synthesized curcumin-based near-infrared (NIR) fluorescence imaging probes for detecting both soluble and insoluble amyloid beta (Aβ) species and then an inhibitor that could attenuate cross-linking of Aβ induced by copper. According to our previous results and the possible structural stereohindrance compatibility of the Aβ peptide and the hydrophobic/hydrophilic property of the Aβ13-20 (HHQKLVFF) fragment, NIR imaging probe CRANAD-58 was designed and synthesized. As expected CRANAD-58 showed significant fluorescence property changes upon mixing with both soluble and insoluble Aβ species in vitro. In vivo NIR imaging revealed that CRANAD-58 was capable of differentiating transgenic and wild-type mice as young as 4 months old, the age that lacks apparently visible Aβ plaques and Aβ is likely in its soluble forms. According to our limited studies on the interaction mechanism between CRANAD-58 and Aβ, we also designed CRANAD-17 to attenuate the cross-linking of Aβ42 induced by copper. It is well-known that the coordination of copper with imidazoles on Histidine-13 and 14 (H13, H14) of Aβ peptides could initialize covalent cross-linking of Aβ. In CRANAD-17, a curcumin scaffold was used as an anchoring moiety to usher the designed compound to the vicinity of H13 and H14 of Aβ, and imidazole rings were incorporated to compete with H13/H14 for copper binding. The results of SDS-PAGE gel and Western blot indicated that CRANAD-17 was capable of inhibiting Aβ42 cross-linking induced by copper. This raises a potential for CRANAD-17 to be considered for AD therapy.
Collapse
Affiliation(s)
- Xueli Zhang
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School , Building 75, Charlestown, Massachusetts 02129, United States
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Prachayasittikul V, Prachayasittikul S, Ruchirawat S, Prachayasittikul V. 8-Hydroxyquinolines: a review of their metal chelating properties and medicinal applications. DRUG DESIGN DEVELOPMENT AND THERAPY 2013; 7:1157-78. [PMID: 24115839 PMCID: PMC3793592 DOI: 10.2147/dddt.s49763] [Citation(s) in RCA: 266] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Metal ions play an important role in biological processes and in metal homeostasis. Metal imbalance is the leading cause for many neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis. 8-Hydroxyquinoline (8HQ) is a small planar molecule with a lipophilic effect and a metal chelating ability. As a result, 8HQ and its derivatives hold medicinal properties such as antineurodegenerative, anticancer, antioxidant, antimicrobial, anti-inflammatory, and antidiabetic activities. Herein, diverse bioactivities of 8HQ and newly synthesized 8HQ-based compounds are discussed together with their mechanisms of actions and structure–activity relationships.
Collapse
Affiliation(s)
- Veda Prachayasittikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Bangkok, Thailand
| | | | | | | |
Collapse
|
42
|
Vranec P, Potočňák I. Low-dimensional compounds containing bioactive ligands. Part VI. Spectral and structural characterizations of a unique zinc(II) compound with biologically active ligand — clioquinol. INORG CHEM COMMUN 2013. [DOI: 10.1016/j.inoche.2013.06.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
43
|
Ayton S, Lei P, Bush AI. Metallostasis in Alzheimer's disease. Free Radic Biol Med 2013; 62:76-89. [PMID: 23142767 DOI: 10.1016/j.freeradbiomed.2012.10.558] [Citation(s) in RCA: 258] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 10/30/2012] [Accepted: 10/30/2012] [Indexed: 12/22/2022]
Abstract
2012 has been another year in which multiple large-scale clinical trials for Alzheimer's disease (AD) have failed to meet their clinical endpoints. With the social and financial burden of this disease increasing every year, the onus is now on the field of AD researchers to investigate alternative ideas to deliver outcomes for patients. Although several major clinical trials targeting Aβ have failed, three smaller clinical trials targeting metal interactions with Aβ have all shown benefit for patients. Here we review the genetic, pathological, biochemical, and pharmacological evidence that underlies the metal hypothesis of AD. The AD-affected brain suffers from metallostasis, or fatigue of metal trafficking, resulting in redistribution of metals into inappropriate compartments. The metal hypothesis is built upon a triad of transition elements: iron, copper, and zinc. The hypothesis has matured from early investigations showing amyloidogenic and oxidative stress consequences of these metals; recently, disease-related proteins, APP, tau, and presenilin, have been shown to have major roles in metal regulation, which provides insight into the pathway of neurodegeneration in AD and illuminates potential new therapeutic avenues.
Collapse
Affiliation(s)
- Scott Ayton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Peng Lei
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Ashley I Bush
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
44
|
Török B, Sood A, Bag S, Tulsan R, Ghosh S, Borkin D, Kennedy AR, Melanson M, Madden R, Zhou W, Levine H, Török M. Diaryl hydrazones as multifunctional inhibitors of amyloid self-assembly. Biochemistry 2013; 52:1137-48. [PMID: 23346953 DOI: 10.1021/bi3012059] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The design and application of an effective, new class of multifunctional small molecule inhibitors of amyloid self-assembly are described. Several compounds based on the diaryl hydrazone scaffold were designed. Forty-four substituted derivatives of this core structure were synthesized using a variety of benzaldehydes and phenylhydrazines and characterized. The inhibitor candidates were evaluated in multiple assays, including the inhibition of amyloid β (Aβ) fibrillogenesis and oligomer formation and the reverse processes, the disassembly of preformed fibrils and oligomers. Because the structure of the hydrazone-based inhibitors mimics the redox features of the antioxidant resveratrol, the radical scavenging effect of the compounds was evaluated by colorimetric assays against 2,2-diphenyl-1-picrylhydrazyl and superoxide radicals. The hydrazone scaffold was active in all of the different assays. The structure-activity relationship revealed that the substituents on the aromatic rings had a considerable effect on the overall activity of the compounds. The inhibitors showed strong activity in fibrillogenesis inhibition and disassembly, and even greater potency in the inhibition of oligomer formation and oligomer disassembly. Supporting the quantitative fluorometric and colorimetric assays, size exclusion chromatographic studies indicated that the best compounds practically eliminated or substantially inhibited the formation of soluble, aggregated Aβ species, as well. Atomic force microscopy was also applied to monitor the morphology of Aβ deposits. The compounds also possessed the predicted antioxidant properties; approximately 30% of the synthesized compounds showed a radical scavenging effect equal to or better than that of resveratrol or ascorbic acid.
Collapse
Affiliation(s)
- Béla Török
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125-3393, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Amir A, Shmuel E, Zagalsky R, Sayer AH, Nadel Y, Fischer B. Nucleoside-5'-phosphorothioate analogues are biocompatible antioxidants dissolving efficiently amyloid beta-metal ion aggregates. Dalton Trans 2012; 41:8539-49. [PMID: 22652964 DOI: 10.1039/c2dt30631j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Amyloid beta (Aβ) peptide is known to precipitate and form aggregates with zinc and copper ions in vitro and, in vivo in Alzheimer's disease (AD) patients. Metal-ion-chelation was suggested as therapy for the metal-ion-induced Aβ aggregation, metal-ion overload, and oxidative stress. In a quest for biocompatible metal-ion chelators potentially useful for AD therapy, we tested a series of nucleoside 5'-phosphorothioate derivatives as re-solubilization agents of Cu(+)/Cu(2+)/Zn(2+)-induced Aβ-aggregates, and inhibitors of Fenton reaction in Cu(+) or Fe(2+)/H(2)O(2) system. The most promising chelator in this series was found to be APCPP-γ-S. This nucleotide was found to be more efficient than EDTA in re-solubilization of Aβ(40)-Cu(2+) aggregates as observed by the lower diameter, d(H), (86 vs. 64 nm, respectively) obtained in dynamic light scattering measurements. Likewise, APCPP-γ-S dissolved Aβ(40)-Cu(+) and Aβ(42)-Cu(2+)/Zn(2+) aggregates, as monitored by (1)H-NMR and turbidity assays, respectively. Furthermore, addition of APCPP-γ-S to nine-day old Aβ(40)-Cu(2+)/Zn(2+) aggregates, resulted in size reduction as observed by transition electron microscopy (diameter reduction from 2.5 to 0.1 μm for Aβ(40)-Cu(2+) aggregates). APCPP-γ-S proved to be more efficient than ascorbic acid and GSH in reducing OH radical production in Fe(2+)/H(2)O(2) system (IC(50) values 85, 216 and, 92 μM, respectively). Therefore, we propose APCPP-γ-S as a potential AD therapy capable of both reducing OH radical production and re-solubilization of Aβ(40/42)-M(n+) aggregates.
Collapse
Affiliation(s)
- Aviran Amir
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | | | | | | | | | | |
Collapse
|
46
|
Török B, Sood A, Bag S, Kulkarni A, Borkin D, Lawler E, Dasgupta S, Landge S, Abid M, Zhou W, Foster M, LeVine H, Török M. Structure-activity relationships of organofluorine inhibitors of β-amyloid self-assembly. ChemMedChem 2012; 7:910-9. [PMID: 22351619 PMCID: PMC4848461 DOI: 10.1002/cmdc.201100569] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 01/30/2012] [Indexed: 12/22/2022]
Abstract
A broad group of structurally diverse small organofluorine compounds were synthesized and evaluated as inhibitors of β-amyloid (Aβ) self-assembly. The main goal was to generate a diverse library of compounds with the same functional group and to observe general structural features that characterize inhibitors of Aβ oligomer and fibril formation, ultimately identifying structures for further focused inhibitor design. The common structural motifs in these compounds are CF(3) -C-OH and CF(3) -C-NH groups that were proposed to be binding units in our previous studies. A broad range of potential small-molecule inhibitors were synthesized by combining various carbocyclic and heteroaromatic rings with an array of substituents, generating a total of 106 molecules. The compounds were tested by standard methods such as thioflavin-T fluorescence spectroscopy for monitoring fibril formation, biotinyl Aβ(1-42) single-site streptavidin-based assays for observing oligomer formation, and atomic force microscopy for morphological studies. These assays revealed a number of structures that show significant inhibition against either Aβ fibril or oligomer formation. A detailed analysis of the structure-activity relationship of anti-fibril and -oligomer properties is provided. These data present further experimental evidence for the distinct nature of fibril versus oligomer formation and indicate that the interaction of the Aβ peptide with chiral small molecules is not stereospecific in nature.
Collapse
Affiliation(s)
- Béla Török
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Abha Sood
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Seema Bag
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Aditya Kulkarni
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Dmitry Borkin
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Elizabeth Lawler
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Sujaya Dasgupta
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Shainaz Landge
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Mohammed Abid
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Weihong Zhou
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Michelle Foster
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| | - Harry LeVine
- Department of Molecular and Cellular Biochemistry, Chandler School of Medicine and Center on Aging, University of Kentucky, Lexington, KY 40536 (USA)
| | - Marianna Török
- Department of Chemistry, University of Massachusetts Boston, 100 Morrissey Blvd, Boston, MA 02125 (USA)
| |
Collapse
|
47
|
Shaw JE, Chio J, Dasgupta S, Lai AY, Mo GCH, Pang F, Thomason LAM, Yang AJ, Yip CM, Nitz M, McLaurin J. Aβ(1-42) assembly in the presence of scyllo-inositol derivatives: identification of an oxime linkage as important for the development of assembly inhibitors. ACS Chem Neurosci 2012; 3:167-77. [PMID: 22860186 DOI: 10.1021/cn2000926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 12/23/2011] [Indexed: 11/29/2022] Open
Abstract
To identify a lead skeleton structure for optimization of scyllo-inositol-based inhibitors of amyloid-beta peptide (Aβ) aggregation, we have synthesized aldoxime, hydroxamate, carbamate, and amide linked scyllo-inositol derivatives. These structures represent backbones that can be readily expanded into a wide array of derivatives. They also provide conservative modifications of the scyllo-inositol backbone, as they maintain the display of the equatorial polar atoms, preserving the stereochemical requirement necessary for maximum inhibition of Aβ(1-42) fiber formation. In addition, a reliable work plan for screening derivatives was developed in order to preferentially identify a backbone(s) structure that prevents fibrillogenesis and stabilizes nontoxic small molecular weight oligomers, as we have previously reported for scyllo-inositol. In the present studies, we have adapted a high throughput ELISA-based oligomerization assay followed by atomic force microscopy to validate the results screen compounds. The lead compounds were then tested for toxicity and ability to rescue Aβ(1-42) induced toxicity in vitro and the affinity of the compounds for Aβ(1-42) compared by mass spectrometry. The data to suggest that compounds must maintain a planar conformation to exhibit activity similar to scyllo-inositol and that the oxime derivative represents the lead backbone for future development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - A. J. Yang
- Department of Anatomy and Neurobiology, University of Maryland at Baltimore, Baltimore, Maryland,
United States
| | | | | | | |
Collapse
|
48
|
Kim SY, Lee MJ, Kim JW, Na YR, Lee HY, Cho H, Lee KB, Lee YM, Lee C, Park H, Yang EG. Effects of Clioquinol Analogues on the Hypoxia-Inducible Factor Pathway and Intracelullar Mobilization of Metal Ions. Biol Pharm Bull 2012. [DOI: 10.1248/bpb.b12-00507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- So Yeon Kim
- Biomedical Research Institute, Korea Institute of Science and Technology
| | - Myong Jin Lee
- Biomedical Research Institute, Korea Institute of Science and Technology
| | - Jeong Won Kim
- Functional Proteomics Center, Korea Institute of Science and Technology
| | - Yu-Ran Na
- Functional Proteomics Center, Korea Institute of Science and Technology
| | - Ho-Youl Lee
- Department of Life Science, University of Seoul
| | - Hyunju Cho
- Biomedical Research Institute, Korea Institute of Science and Technology
| | - Keun Byeol Lee
- Department of Natural Sciences, Kyungpook National University
| | - You Mie Lee
- Department of Natural Sciences, Kyungpook National University
| | - Cheolju Lee
- Biomedical Research Institute, Korea Institute of Science and Technology
| | | | - Eun Gyeong Yang
- Biomedical Research Institute, Korea Institute of Science and Technology
- Functional Proteomics Center, Korea Institute of Science and Technology
| |
Collapse
|
49
|
Li G, Rauscher S, Baud S, Pomès R. Binding of inositol stereoisomers to model amyloidogenic peptides. J Phys Chem B 2011; 116:1111-9. [PMID: 22091989 DOI: 10.1021/jp208567n] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The self-aggregation of proteins into amyloid fibrils is a pathological hallmark of numerous incurable diseases such as Alzheimer's disease. scyllo-Inositol is a stereochemistry-dependent in vitro inhibitor of amyloid formation. As the first step to elucidate its mechanism of action, we present molecular dynamics simulations of scyllo-inositol and its inactive stereoisomer, chiro-inositol, with simple peptide models, alanine dipeptide (ADP) and (Gly-Ala)(4). We characterize molecular interactions and compute equilibrium binding constants between inositol and ADP as well as, successively, monomers, amorphous aggregates, and fibril-like β-sheet aggregates of (Gly-Ala)(4). Inositol interacts weakly with all peptide systems considered, with millimolar to molar affinities, and displaces the conformational equilibria of ADP but not of the (Gly-Ala)(4) systems. However, scyllo- and chiro-inositol adopt different binding modes on the surface of β-sheet aggregates. These results suggest that inositol does not inhibit amyloid formation by breaking up preformed aggregates but rather by binding to the surface of prefibrillar aggregates.
Collapse
Affiliation(s)
- Grace Li
- Department of Biochemistry, University of Toronto, 27 King's College Circle, Toronto, Ontario, Canada M5S 1A1
| | | | | | | |
Collapse
|
50
|
LeVine H, Lampe L, Abdelmoti L, Augelli-Szafran CE. Dihydroxybenzoic acid isomers differentially dissociate soluble biotinyl-Aβ(1-42) oligomers. Biochemistry 2011; 51:307-15. [PMID: 22129351 DOI: 10.1021/bi201288x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Polyphenolic compounds including a number of natural products such as resveratrol, curcumin, catechin derivatives, and nordihydroguaiaretic acid have effects on the assembly of Aβ fibrils and oligomers as well as on fibril morphology. Based on a lead structure obtained from a screen of a small molecule diversity library, simple benzoic acid derivatives distinguished by the number and position of hydroxyls on the aromatic ring displayed different abilities to dissociate preformed biotinyl-Aβ(1-42) oligomers. The 2,3-, 2,5-, and 3,4-dihydroxybenzoic acid (DHBA) isomers were active oligomer dissociators. The remaining DHBA isomers and the monohydroxy and unsubstituted benzoic acids were inactive and did not compete with the active compounds to block oligomer dissociation. None of the compounds blocked oligomer assembly, indicating that they do not interact with monomeric Aβ to shift the oligomer-monomer equilibrium. Dissociating activity was not associated with quinone redox cycling capacity of the compounds. Gallic acid (3,4,5-trihydroxybenzoic acid) stabilized biotinyl-Aβ(1-42) oligomers against intrinsic dissociation and blocked the effects of the active dissociators, independent of the concentration of dissociator. A model for the mechanism of action of the DHBA dissociators proposes that these compounds destabilize oligomer structure promoting progressive monomer dissociation rather than fissioning oligomers into smaller, but still macromolecular, species. Gallic acid blocks dissociation by stabilizing oligomers against this process.
Collapse
Affiliation(s)
- Harry LeVine
- Department of Cellular and Molecular Biochemistry, Center on Aging, Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40506, United States.
| | | | | | | |
Collapse
|