1
|
Jawad M, Uthirapathy S, Altalbawy FMA, Oghenemaro EF, Rizaev J, Lal M, Eldesoqui M, Sharma N, Pramanik A, Al-Hamairy AK. Examining the role of antioxidant supplementation in mitigating oxidative stress markers in Alzheimer's disease: a comprehensive review. Inflammopharmacology 2024:10.1007/s10787-024-01622-9. [PMID: 39699843 DOI: 10.1007/s10787-024-01622-9] [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: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/20/2024]
Abstract
Alzheimer's disease is a devastating neurodegenerative disorder that affects millions of people worldwide. One of the key pathological features of Alzheimer's disease is oxidative stress, which is characterized by an imbalance between the production of reactive oxygen species and the body's ability to neutralize them with antioxidants. In recent years, there has been growing interest in the potential role of antioxidant supplementation in mitigating oxidative stress markers in Alzheimer's disease. This review paper aims to provide a comprehensive overview of the current research on antioxidant supplementation in Alzheimer's disease and its effects on oxidative stress markers. The paper will examine the underlying mechanisms of oxidative stress in Alzheimer's disease, the potential benefits of antioxidant supplementation, and the challenges and limitations of using antioxidants as a therapeutic strategy.
Collapse
Affiliation(s)
- Mahmood Jawad
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Subasini Uthirapathy
- Pharmacology Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, PMB 1, Abraka, Delta State, Nigeria
| | - Jasur Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Madan Lal
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah, 13713, Riyadh, Saudi Arabia.
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Ahmed Khudhair Al-Hamairy
- Anesthesia Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, Babylon, Iraq
| |
Collapse
|
2
|
Behar AE, Maayan G. A cocktail of Cu 2+- and Zn 2+-peptoid-based chelators can stop ROS formation for Alzheimer's disease therapy. Chem Sci 2024:d4sc04313h. [PMID: 39464602 PMCID: PMC11503657 DOI: 10.1039/d4sc04313h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
The formation of reactive oxygen species (ROS) in the brain is a major cause of neuropathologic degradation associated with Alzheimer's Disease (AD). It has been suggested that the copper (Cu)-amyloid-β (Aβ) peptide complex can lead to ROS formation in the brain. An external chelator for Cu that can extract Cu from the CuAβ complex should inhibit the formation of ROS, making Cu chelation an excellent therapeutic approach for AD. Such a chelator should possess high selectivity for Cu over zinc (Zn), which is also present within the synaptic cleft. However, such selectivity is generally hard to achieve in one molecule due to the similarities in the binding preferences of these two metal ions. As an alternative to monotherapy (where Cu extraction is performed using a single chelator), herein we describe a variation of combination therapy - a novel cocktail approach, which is based on the co-administration of two structurally different peptidomimetic chelators, aiming to target both Cu2+ and Zn2+ ions simultaneously but independently from each other. Based on rigorous spectroscopic experiments, we demonstrate that our peptidomimetic cocktail allows, for the first time, the complete and immediate inhibition of ROS production by the CuAβ complex in the presence of Zn2+. In addition, we further demonstrate the high stability of the cocktail under simulated physiological conditions and its resistance to proteolytic degradation by trypsin and report the water/n-octanol partition coefficient, initially assessing the blood-brain barrier (BBB) permeability potential of the chelators.
Collapse
Affiliation(s)
- Anastasia E Behar
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City 3200008 Haifa Israel
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion - Israel Institute of Technology, Technion City 3200008 Haifa Israel
| |
Collapse
|
3
|
Gómez-Castro CZ, Quintanar L, Vela A. An N-terminal acidic β-sheet domain is responsible for the metal-accumulation properties of amyloid-β protofibrils: a molecular dynamics study. J Biol Inorg Chem 2024; 29:407-425. [PMID: 38811408 PMCID: PMC11186886 DOI: 10.1007/s00775-024-02061-1] [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: 12/20/2023] [Accepted: 04/10/2024] [Indexed: 05/31/2024]
Abstract
The influence of metal ions on the structure of amyloid- β (Aβ) protofibril models was studied through molecular dynamics to explore the molecular mechanisms underlying metal-induced Aβ aggregation relevant in Alzheimer's disease (AD). The models included 36-, 48-, and 188-mers of the Aβ42 sequence and two disease-modifying variants. Primary structural effects were observed at the N-terminal domain, as it became susceptible to the presence of cations. Specially when β-sheets predominate, this motif orients N-terminal acidic residues toward one single face of the β-sheet, resulting in the formation of an acidic region that attracts cations from the media and promotes the folding of the N-terminal region, with implications in amyloid aggregation. The molecular phenotype of the protofibril models based on Aβ variants shows that the AD-causative D7N mutation promotes the formation of N-terminal β-sheets and accumulates more Zn2+, in contrast to the non-amyloidogenic rodent sequence that hinders the β-sheets and is more selective for Na+ over Zn2+ cations. It is proposed that forming an acidic β-sheet domain and accumulating cations is a plausible molecular mechanism connecting the elevated affinity and concentration of metals in Aβ fibrils to their high content of β-sheet structure at the N-terminal sequence.
Collapse
Affiliation(s)
- Carlos Z Gómez-Castro
- Conahcyt-Universidad Autónoma del Estado de Hidalgo, Km 4.5 Carr. Pachuca-Tulancingo, Mineral de La Reforma, 42184, Hidalgo, Mexico.
| | - Liliana Quintanar
- Department of Chemistry, Cinvestav, Av. Instituto Politécnico Nacional 2508, CDMX, San Pedro Zacatenco, 07360, Gustavo A. Madero, Mexico.
| | - Alberto Vela
- Department of Chemistry, Cinvestav, Av. Instituto Politécnico Nacional 2508, CDMX, San Pedro Zacatenco, 07360, Gustavo A. Madero, Mexico.
| |
Collapse
|
4
|
Rulmont C, Stigliani JL, Hureau C, Esmieu C. Rationally Designed Cu(I) Ligand to Prevent CuAβ-Generated ROS Production in the Alzheimer's Disease Context. Inorg Chem 2024; 63:2340-2351. [PMID: 38243896 DOI: 10.1021/acs.inorgchem.3c02693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
In the context of Alzheimer's disease, copper (Cu) can be loosely bound to the amyloid-β (Aβ) peptide, leading to the formation of CuAβ, which can catalytically generate reactive oxygen species that contribute to oxidative stress. To fight against this phenomenon, the chelation therapy approach has been developed and consists of using a ligand able to remove Cu from Aβ and to redox-silence it, thus stopping the reactive oxygen species (ROS) production. A large number of Cu(II) chelators has been studied, allowing us to define and refine the properties required to design a "good" ligand, but without strong therapeutic outcomes to date. Those chelators targeted the Cu(II) redox state. Herein, we explore a parallel and relevant alternative pathway by designing a chelator able to target the Cu(I) redox state. To that end, we designed LH2 ([1N3S] binding set) and demonstrated that (i) it is perfectly able to extract Cu(I) from Cu(I)Aβ even in the presence of an excess of Zn(II) and (ii) it redox-silences the Cu, preventing the formation of ROS. We showed that LH2 that is sensitive to oxidation can efficiently replace the [Zn(II)L] complex without losing its excellent ability to stop the ROS production while increasing its resistance to oxidation.
Collapse
Affiliation(s)
- Clément Rulmont
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse 31077, France
| | | | | | - Charlène Esmieu
- LCC-CNRS, Université de Toulouse, CNRS, Toulouse 31077, France
| |
Collapse
|
5
|
Sohrabi M, Bozorgmehr MR, Momen-Heravi M. Investigating the combined effect of copper, zinc, and iron ions on truncated and full-length Aβ peptides: insights from molecular dynamics simulation. J Biomol Struct Dyn 2024:1-9. [PMID: 38189361 DOI: 10.1080/07391102.2024.2301755] [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: 09/23/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
The truncated Aβ1 - 16 peptide containing the metal-binding domain is frequently used in in silico and experimental investigations because it is more soluble and thus more suitable for studies in solution and does not form amyloids. Several spectroscopic studies have shown that the metal binding of Aβ1 - 16 is very similar to that of the full-length Aβ1 - 42. However, since small changes can have a significant impact on aggregation, further experimental and theoretical are needed to elucidate the detailed structures of truncated and full-length Aβ. In this research, the binding of copper ion to the Aβ1 - 16 and Aβ1 - 42 has been studied by molecular dynamics simulation method. To investigate the effect of copper ion on beta-amyloid peptide structure, the simulations were repeated in the copper and zinc ions, copper and iron binary system, and the copper, zinc and iron ions ternary system. The conformation factor was calculated to calculate the binding affinity of copper ion to beta-amyloid peptide residues. The results showed that the initial 16 residues of the beta-amyloid peptide have high binding affinity for copper ions, and histidine 13 and histidine 14 have significantly higher binding affinity for copper ions in all studied systems. Zinc and iron ions were found to reduce the conformational factor of peptide residues in binding to copper ions, and the aggregation tendency was lower in the truncated structure. The SASA results suggest that the side chains of peptide residues are more affected by shortening and the presence of ions.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mona Sohrabi
- Department of Chemistry, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | | |
Collapse
|
6
|
Kepp KP, Robakis NK, Høilund-Carlsen PF, Sensi SL, Vissel B. The amyloid cascade hypothesis: an updated critical review. Brain 2023; 146:3969-3990. [PMID: 37183523 DOI: 10.1093/brain/awad159] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/16/2023] Open
Abstract
Results from recent clinical trials of antibodies that target amyloid-β (Aβ) for Alzheimer's disease have created excitement and have been heralded as corroboration of the amyloid cascade hypothesis. However, while Aβ may contribute to disease, genetic, clinical, imaging and biochemical data suggest a more complex aetiology. Here we review the history and weaknesses of the amyloid cascade hypothesis in view of the new evidence obtained from clinical trials of anti-amyloid antibodies. These trials indicate that the treatments have either no or uncertain clinical effect on cognition. Despite the importance of amyloid in the definition of Alzheimer's disease, we argue that the data point to Aβ playing a minor aetiological role. We also discuss data suggesting that the concerted activity of many pathogenic factors contribute to Alzheimer's disease and propose that evolving multi-factor disease models will better underpin the search for more effective strategies to treat the disease.
Collapse
Affiliation(s)
- Kasper P Kepp
- Section of Biophysical and Biomedicinal chemistry, DTU Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Nikolaos K Robakis
- Icahn School of Medicine at Mount Sinai Medical Center, New York, NY 10029, USA
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Stefano L Sensi
- Center for Advanced Studies and Technology-CAST, and Institute for Advanced Biotechnology (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
| | - Bryce Vissel
- St Vincent's Hospital Centre for Applied Medical Research, St Vincent's Hospital, Sydney, 2010, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, Sydney, NSW 2052, Australia
| |
Collapse
|
7
|
Abelein A. Metal Binding of Alzheimer's Amyloid-β and Its Effect on Peptide Self-Assembly. Acc Chem Res 2023; 56:2653-2663. [PMID: 37733746 PMCID: PMC10552549 DOI: 10.1021/acs.accounts.3c00370] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 09/23/2023]
Abstract
Metal ions have been identified as key factors modulating the aggregation of amyloid-β peptide (Aβ) implicated in Alzheimer's disease (AD). The presence of elevated levels of metal ions in the amyloid plaques in AD patients supports the notion that the dysfunction of metal homeostasis is connected to the development of AD pathology. Here, recent findings from high- and low-resolution biophysical techniques are put into perspective, providing detailed insights into the molecular structures and dynamics of metal-bound Aβ complexes and the effect of metal ions on the Aβ aggregation process. In particular, the development of theoretical kinetic models deducing different microscopic nucleation events from the macroscopic aggregation behavior has enabled deciphering of the effect of metal ions on specific nucleation processes. In addition to these macroscopic measurements of bulk aggregation to quantify microscopic rates, recent NMR studies have revealed details about the structures and dynamics of metal-Aβ complexes, thereby linking structural events to bulk aggregation. Interestingly, transition-metal ions, such as copper, zinc, and silver ions, form a compact complex with the N-terminal part of monomeric Aβ, respectively, where the metal-bound "folded" state is in dynamic equilibrium with an "unfolded" state. The rates and thermodynamic features of these exchange dynamics have been determined by using NMR relaxation dispersion experiments. Additionally, the application of specifically tailored paramagnetic NMR experiments on the Cu(II)-Aβ complex has been fruitful in obtaining structural constraints within the blind sphere of conventional NMR experiments. This enables the determination of molecular structures of the "folded" Cu(II)-coordinated N-terminal region of Aβ. Furthermore, the discussed transition-metal ions modulate Aβ self-assembly in a concentration-dependent manner, where low metal ion concentrations inhibit Aβ fibril formation, while at high metal ion concentrations other processes occur, resulting in amorphous aggregate formation. Remarkably, the metal-Aβ interaction predominately reduces one specific nucleation step, the fibril-end elongation, whereas primary and surface-catalyzed secondary nucleation mechanisms are less affected. Specific inhibition of fibril-end elongation theoretically predicts an enhanced generation of Aβ oligomers, which is an interesting contribution to understanding metal-Aβ-associated neurotoxic effects. Taken together, the metal binding process creates a metal-bound Aβ complex, which is seemingly inert to aggregation. This process hence efficiently reduces the aggregation-prone peptide pool, which on the macroscopic level is reflected as slower aggregation kinetics. Thus, the specific binding of metals to the Aβ monomer can be linked to the macroscopic inhibitory effect on Aβ bulk aggregation, providing a molecular understanding of the Aβ aggregation mechanism in the presence of metal ions, where the metal ion can be seen as a minimalist agent against Aβ self-assembly. These insights can help to target Aβ aggregation in vivo, where metal ions are key factors modulating the Aβ self-assembly and Aβ-associated neurotoxicity.
Collapse
Affiliation(s)
- Axel Abelein
- Department of Biosciences
and Nutrition, Karolinska Institutet, 141 52 Huddinge, Sweden
| |
Collapse
|
8
|
Feng J, She Y, Li C, Shen L. Metal ion mediated aggregation of Alzheimer's disease peptides and proteins in solutions and at surfaces. Adv Colloid Interface Sci 2023; 320:103009. [PMID: 37776735 DOI: 10.1016/j.cis.2023.103009] [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: 06/20/2023] [Revised: 08/29/2023] [Accepted: 09/24/2023] [Indexed: 10/02/2023]
Abstract
Although the pathogenesis of Alzheimer's disease (AD) is still unclear, abnormally high concentrations of metal ions, like copper, iron and zinc, were found in senile plaques of AD brain, which inspires extensive studies on the fundamental molecular interactions of metal ions with the pathogenic hallmarks, amyloid-β (Aβ) peptides and tau proteins, respectively forming senile plaques and neurofibrillary tangles (NFTs) in AD brains. Early works concern the concentration effect of the metal ions on Aβ and tau aggregation. Yet, it is obvious that the surrounding environment of the metal ions must also be considered, not just the metal ions as free accessible forms in the solution phase. The most important surrounding environment in vivo is a very large surface area from cell membranes and other macromolecular surfaces. These bio-interfaces make the kinetic pathways of metal ion mediated Aβ and tau aggregation radically different from those in the solution phase. To better understand the role of metal ions in AD peptide and protein aggregation, we summarize and discuss the recent achievements in the research of metal ion mediated Aβ and tau aggregation, particularly the corresponding mechanism differences between the solution phase and the surface environment. The metal ion chelation therapy for AD is also discussed from the point of the surface pool of metal ions.
Collapse
Affiliation(s)
- Jiahao Feng
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Yifei She
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Chongjia Li
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China
| | - Lei Shen
- Key Laboratory for Neurodegenerative Diseases Nanomedicine of Hubei Province, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China.
| |
Collapse
|
9
|
Shen H, Dou Y, Wang X, Wang X, Kong F, Wang S. Guluronic acid can inhibit copper(II) and amyloid - β peptide coordination and reduce copper-related reactive oxygen species formation associated with Alzheimer's disease. J Inorg Biochem 2023; 245:112252. [PMID: 37207465 DOI: 10.1016/j.jinorgbio.2023.112252] [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: 02/07/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
Copper-related reactive oxygen species (ROS) formation can lead to neuropathologic degradation associated with Alzheimer's disease (AD) according to amyloid cascade hypothesis. A complexing agent that can selectively chelate with copper ions and capture copper ions from the complex formed by copper ions and amyloid-β (Cu - Aβ complex) may be available in reducing ROS formation. Herein, we described applications of guluronic acid (GA), a natural oligosaccharide complexing agent obtained from enzymatic hydrolysis of brown algae, in reducing copper-related ROS formation. UV-vis absorption spectra demonstrated the coordination between GA and Cu(II). Ascorbic acid consumption and coumarin-3-carboxylic acid fluorescence assays confirmed the viability of GA in reducing ROS formation in solutions containing other metal ions and Aβ. Fluorescence kinetics, DPPH radical clearance and high resolution X - ray photoelectron spectroscopy results revealed the reductivity of GA. Human liver hepatocellular carcinoma (HepG2) cell viability demonstrated the biocompatibility of GA at concentrations lower than 320 μM. Cytotoxic results of human neuroblastoma (SH-SY5Y) cells verified that GA can inhibit copper-related ROS damage in neuronal cells. Our findings, combined with the advantages of marine drugs, make GA a promising candidate in reducing copper-related ROS formation associated with AD therapy.
Collapse
Affiliation(s)
- Hangyu Shen
- State Key Laboratory of Biobased Materials and Green Papermaking, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong 250353, PR China
| | - Yun Dou
- State Key Laboratory of Biobased Materials and Green Papermaking, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong 250353, PR China
| | - Xiaoying Wang
- State Key Laboratory of Biobased Materials and Green Papermaking, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong 250353, PR China.
| | - Xiaohui Wang
- State Key Laboratory of Biobased Materials and Green Papermaking, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong 250353, PR China
| | - Fangong Kong
- State Key Laboratory of Biobased Materials and Green Papermaking, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong 250353, PR China
| | - Shoujuan Wang
- State Key Laboratory of Biobased Materials and Green Papermaking, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong 250353, PR China.
| |
Collapse
|
10
|
Poore AT, Zuercher EC, Bury G, Whitesell C, Nguyen CC, Pushkar YN, Tian S. Revisit the E2 Domain of Amyloid Precursor Protein: Ferroxidase, Superoxide and Peroxynitrite Scavenging Activities. Inorg Chem 2023. [PMID: 37369063 DOI: 10.1021/acs.inorgchem.3c01336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023]
Abstract
Amyloid precursor protein (APP) is the biological precursor of β-amyloids, a known histopathological hallmark associated with Alzheimer's disease (AD). The function of APP is of great interest yet remains elusive. One of the extracellular domains of APP, the E2 domain, has been proposed to possess ferroxidase activity and affect neuronal iron homeostasis. However, contradicting evidence has been reported, and its precise role remains inconclusive. Here, we studied the Cu-binding site of the E2 domain using extended X-ray absorption fine structure (EXAFS), UV-vis, and electron paramagnetic resonance (EPR) and discovered that a new labile water ligand coordinates to the Cu(II) cofactor in addition to the four known histidines. We explored the proposed ferroxidase activity of the Cu(II)-E2 domain through reactions with ferrous iron and observed single-turnover ferrous oxidation activity with a rate up to 1.0 × 102 M-1 s-1. Cu(I)-E2 reacted with molecular oxygen at a rate of only 5.3 M-1 s-1, which would restrict any potential multiturnover ferroxidase activity to this slow rate and prevents observation of activity under multiturnover conditions. The positive electrostatic potential surface of the protein indicates possible reactivity with negatively charged small substrates such as superoxide radicals (O2•-) and peroxynitrite (ONOO-) that are major contributors to the oxidative stress prevalent in the extracellular environment. Our assays showed that Cu(I)-E2 can remove O2•- at a rate of 1.6 × 105 M-1 s-1, which is slower than the rates of native SODs. However, the reaction between Cu(I)-E2 and ONOO- achieved a rate of 1.1 × 105 M-1 s-1, comparable to native ONOO- scavenger peroxiredoxins (105-107 M-1 s-1). Therefore, the E2 domain of APP can serve as an enzymatic site that may function as a ferroxidase under substrate-limiting conditions, a supplemental O2•- scavenger, and an ONOO- remover in the vicinity of the cellular iron efflux channel and protect neuron cells from reactive oxygen species (ROS) and reactive nitrogen species (RNS) damage.
Collapse
Affiliation(s)
- Andrew T Poore
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, United States
| | - Eli C Zuercher
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, United States
| | - Gabriel Bury
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Caslyn Whitesell
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, United States
| | - Cuong C Nguyen
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, United States
| | - Yulia N Pushkar
- Department of Physics and Astronomy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shiliang Tian
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47906, United States
| |
Collapse
|
11
|
Malikidogo KP, Drommi M, Atrián-Blasco E, Hormann J, Kulak N, Esmieu C, Hureau C. Ability of Azathiacyclen Ligands To Stop Cu(Aβ)-Induced Production of Reactive Oxygen Species: [3N1S] Is the Right Donor Set. Chemistry 2023; 29:e202203667. [PMID: 36606721 DOI: 10.1002/chem.202203667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/05/2023] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease that leads to the progressive and irreversible loss of mental functions. The amyloid beta (Aβ) peptide involved in the disease is responsible for the production of damaging reactive oxygen species (ROS) when bound to Cu ions. A therapeutic approach that consists of removing Cu ions from Aβ to alter this deleterious interaction is currently being developed. In this context, we report the ability of five different 12-membered thiaazacyclen ligands to capture Cu from Aβ and to redox silence it. We propose that the presence of a sole sulfur atom in the ligand increases the rate of Cu capture and removal from Aβ, while the kinetic aspect of the chelation was an issue encountered with the 4N parent ligand. The best ligand for removing Cu from Aβ and inhibiting the associated ROS production is the 1-thia-4,7,10-triazacyclododecane [3N1S]. Indeed the replacement of more N by S atoms makes the corresponding Cu complexes easier to reduce and thus able to produce ROS on their own. In addition, the ligand with three sulfur atoms has a weaker affinity for CuII than Aβ, and is thus unable to remove Cu from CuAβ.
Collapse
Affiliation(s)
- Kyangwi P Malikidogo
- LCC-CNRS, Université de Toulouse, CNRS, 31400, Toulouse, France.,Université Grenoble Alpes, DCM (UMR 5250) - CNRS and CEA, IRIG, LCBM (UMR, 5249, Grenoble, France
| | - Marielle Drommi
- LCC-CNRS, Université de Toulouse, CNRS, 31400, Toulouse, France
| | - Elena Atrián-Blasco
- LCC-CNRS, Université de Toulouse, CNRS, 31400, Toulouse, France.,Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Zaragoza, 50009, Spain
| | - Jan Hormann
- Institut für Chemie und Biochemie, Freie Universität Berlin, Fabeckstr. 34/36, 14195, Berlin, Germany
| | - Nora Kulak
- Institut für Chemie und Biochemie, Freie Universität Berlin, Fabeckstr. 34/36, 14195, Berlin, Germany.,Institut für Chemie, Otto-von-Guericke-Universität Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Charlène Esmieu
- LCC-CNRS, Université de Toulouse, CNRS, 31400, Toulouse, France
| | | |
Collapse
|
12
|
Yi Y, Lim MH. Current understanding of metal-dependent amyloid-β aggregation and toxicity. RSC Chem Biol 2023; 4:121-131. [PMID: 36794021 PMCID: PMC9906324 DOI: 10.1039/d2cb00208f] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022] Open
Abstract
The discovery of effective therapeutics targeting amyloid-β (Aβ) aggregates for Alzheimer's disease (AD) has been very challenging, which suggests its complicated etiology associated with multiple pathogenic elements. In AD-affected brains, highly concentrated metals, such as copper and zinc, are found in senile plaques mainly composed of Aβ aggregates. These metal ions are coordinated to Aβ and affect its aggregation and toxicity profiles. In this review, we illustrate the current view on molecular insights into the assembly of Aβ peptides in the absence and presence of metal ions as well as the effect of metal ions on their toxicity.
Collapse
Affiliation(s)
- Yelim Yi
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
13
|
Sarlet A, Ruffine V, Blank KG, Bidan CM. Influence of Metal Cations on the Viscoelastic Properties of Escherichia coli Biofilms. ACS OMEGA 2023; 8:4667-4676. [PMID: 36777596 PMCID: PMC9910073 DOI: 10.1021/acsomega.2c06438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
Biofilms frequently cause complications in various areas of human life, e.g., in medicine and in the food industry. More recently, biofilms are discussed as new types of living materials with tunable mechanical properties. In particular, Escherichia coli produces a matrix composed of amyloid-forming curli and phosphoethanolamine-modified cellulose fibers in response to suboptimal environmental conditions. It is currently unknown how the interaction between these fibers contributes to the overall mechanical properties of the formed biofilms and if extrinsic control parameters can be utilized to manipulate these properties. Using shear rheology, we show that biofilms formed by the E. coli K-12 strain AR3110 stiffen by a factor of 2 when exposed to the trivalent metal cations Al(III) and Fe(III), while no such response is observed for the bivalent cations Zn(II) and Ca(II). Strains producing only one matrix component did not show any stiffening response to either cation or even a small softening. No stiffening response was further observed when strains producing only one type of fiber were co-cultured or simply mixed after biofilm growth. These results suggest that the E. coli biofilm matrix is a uniquely structured composite material when both matrix fibers are produced from the same bacterium. While the exact interaction mechanism between curli, phosphoethanolamine-modified cellulose, and trivalent metal cations is currently not known, our results highlight the potential of using extrinsic parameters to understand and control the interplay between biofilm structure and mechanical properties. This will ultimately aid in the development of better strategies for controlling biofilm growth.
Collapse
Affiliation(s)
- Adrien Sarlet
- Department
of Biomaterials, Max Planck Institute of
Colloids and Interfaces, Am Mühlenberg 1, 14476Potsdam, Germany
| | - Valentin Ruffine
- Mechano(bio)chemistry, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476Potsdam, Germany
| | - Kerstin G. Blank
- Mechano(bio)chemistry, Max Planck Institute of Colloids and Interfaces, Am Mühlenberg 1, 14476Potsdam, Germany
- Institute
of Experimental Physics, Johannes Kepler
University, Altenberger
Str. 69, 4040Linz, Austria
| | - Cécile M. Bidan
- Department
of Biomaterials, Max Planck Institute of
Colloids and Interfaces, Am Mühlenberg 1, 14476Potsdam, Germany
| |
Collapse
|
14
|
Li ZW, Tan B, Wu ZF, Huang XY. A Robust Strontium Coordination Polymer with Selective and Sensitive Fluorescence Sensing Ability for Fe 3+ Ions. MATERIALS (BASEL, SWITZERLAND) 2023; 16:577. [PMID: 36676316 PMCID: PMC9866177 DOI: 10.3390/ma16020577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 06/17/2023]
Abstract
Exploration of sensitive and selective fluorescence sensors towards toxic metal species is of great importance to solve metal pollution issues. In this work, a three-dimensional (3D) strontium coordination polymer of Sr2(tcbpe) (H4tcbpe = 1,1,2,2-tetrakis(4-(4-carboxy-phenyl)phenyl)ethene) has been synthesized and developed as a fluorescent sensor to Fe3+ ions. Sr2(tcbpe) shows a mechanochromic fluorescence with emission shifting from blue of the pristine to green after being ground. Notably, based on a fluorescence quenching mechanism, Sr2(tcbpe) displays a sensitive and selective fluorescent sensing behavior to Fe3+ ions with a detection limit of 0.14 mM. Moreover, Sr2(tcbpe) exhibits high tolerance to water in a wide pH range (pH = 3-13), demonstrating that Sr2(tcbpe) is a potential fluorescent sensor of Fe3+ in water.
Collapse
Affiliation(s)
- Zi-Wei Li
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, The Chinese Academy of Sciences, Fuzhou 350002, China
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China
| | - Bin Tan
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, The Chinese Academy of Sciences, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350108, China
| | - Zhao-Feng Wu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, The Chinese Academy of Sciences, Fuzhou 350002, China
- Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350108, China
| | - Xiao-Ying Huang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, The Chinese Academy of Sciences, Fuzhou 350002, China
| |
Collapse
|
15
|
Sequence-Activity Relationship of ATCUN Peptides in the Context of Alzheimer's Disease. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227903. [PMID: 36432004 PMCID: PMC9698028 DOI: 10.3390/molecules27227903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/17/2022]
Abstract
Amino-terminal CuII and NiII (ATCUN) binding sequences are widespread in the biological world. Here, we report on the study of eight ATCUN peptides aimed at targeting copper ions and stopping the associated formation of reactive oxygen species (ROS). This study was actually more focused on Cu(Aβ)-induced ROS production in which the Aβ peptide is the "villain" linked to Alzheimer's disease. The full characterization of CuII binding to the ATCUN peptides, the CuII extraction from CuII(Aβ), and the ability of the peptides to prevent and/or stop ROS formation are described in the relevant biological conditions. We highlighted in this research that all the ATCUN motifs studied formed the same thermodynamic complex but that the addition of a second histidine in position 1 or 2 allowed for an improvement in the CuII uptake kinetics. This kinetic rate was directly related to the ability of the peptide to stop the CuII(Aβ)-induced production of ROS, with the most efficient motifs being HWHG and HGHW.
Collapse
|
16
|
Gonzalez P, Sabater L, Mathieu E, Faller P, Hureau C. Why the Ala-His-His Peptide Is an Appropriate Scaffold to Remove and Redox Silence Copper Ions from the Alzheimer's-Related Aβ Peptide. Biomolecules 2022; 12:1327. [PMID: 36291536 PMCID: PMC9599918 DOI: 10.3390/biom12101327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
The progressive, neurodegenerative Alzheimer's disease (AD) is the most widespread dementia. Due to the ageing of the population and the current lack of molecules able to prevent or stop the disease, AD will be even more impactful for society in the future. AD is a multifactorial disease, and, among other factors, metal ions have been regarded as potential therapeutic targets. This is the case for the redox-competent Cu ions involved in the production of reactive oxygen species (ROS) when bound to the Alzheimer-related Aβ peptide, a process that contributes to the overall oxidative stress and inflammation observed in AD. Here, we made use of peptide ligands to stop the Cu(Aβ)-induced ROS production and we showed why the AHH sequence is fully appropriate, while the two parents, AH and AAH, are not. The AHH peptide keeps its beneficial ability against Cu(Aβ)-induced ROS, even in the presence of ZnII-competing ions and other biologically relevant ions. The detailed kinetic mechanism by which AHH could exert its action against Cu(Aβ)-induced ROS is also proposed.
Collapse
Affiliation(s)
- Paulina Gonzalez
- LCC-CNRS, Université de Toulouse, CNRS, 31077 Toulouse, France
- Laboratory of Biometals and Biological Chemistry, Institut de Chimie (UMR 7177), Université de Strasbourg-CNRS, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | - Laurent Sabater
- LCC-CNRS, Université de Toulouse, CNRS, 31077 Toulouse, France
| | - Emilie Mathieu
- LCC-CNRS, Université de Toulouse, CNRS, 31077 Toulouse, France
| | - Peter Faller
- Laboratory of Biometals and Biological Chemistry, Institut de Chimie (UMR 7177), Université de Strasbourg-CNRS, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | | |
Collapse
|
17
|
Yang H, Zeng F, Luo Y, Zheng C, Ran C, Yang J. Curcumin Scaffold as a Multifunctional Tool for Alzheimer's Disease Research. Molecules 2022; 27:3879. [PMID: 35745002 PMCID: PMC9227459 DOI: 10.3390/molecules27123879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative disorders, which is caused by multi-factors and characterized by two histopathological hallmarks: amyloid-β (Aβ) plaques and neurofibrillary tangles of Tau proteins. Thus, researchers have been devoting tremendous efforts to developing and designing new molecules for the early diagnosis of AD and curative purposes. Curcumin and its scaffold have fluorescent and photochemical properties. Mounting evidence showed that curcumin scaffold had neuroprotective effects on AD such as anti-amyloidogenic, anti-inflammatory, anti-oxidative and metal chelating. In this review, we summarized different curcumin derivatives and analyzed the in vitro and in vivo results in order to exhibit the applications in AD diagnosis, therapeutic monitoring and therapy. The analysis results showed that, although curcumin and its analogues have some disadvantages such as short wavelength and low bioavailability, these shortcomings can be conquered by modifying the structures. Curcumin scaffold still has the potential to be a multifunctional tool for AD research, including AD diagnosis and therapy.
Collapse
Affiliation(s)
- Haijun Yang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China; (H.Y.); (Y.L.)
| | - Fantian Zeng
- School of Public Health, Xiamen University, Xiamen 361000, China;
| | - Yunchun Luo
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China; (H.Y.); (Y.L.)
| | - Chao Zheng
- PET Center, School of Medicine, Yale University, New Haven, CT 06520, USA;
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Jian Yang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China; (H.Y.); (Y.L.)
| |
Collapse
|
18
|
Chauhan BS, Rai A, Sonkar AK, Tripathi K, Upadhyay S, Mishra L, Srikrishna S. Neuroprotective Activity of a Novel Synthetic Rhodamine-Based Hydrazone against Cu 2+-Induced Alzheimer's Disease in Drosophila. ACS Chem Neurosci 2022; 13:1566-1579. [PMID: 35476931 DOI: 10.1021/acschemneuro.2c00144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
A new rhodamine-based probe 3,5-di-tert-butylsalicylaldehyde rhodamine hydrazone (RHTB) has been synthesized and well characterized using spectroscopic techniques and single-crystal X-ray crystallography. Among several metal ions, it selectively detects Cu2+ ions as monitored by UV-Vis and emission spectral titrations. It displays "turn on" behavior owing to the opening of a spirolactum ring and the presence of 3,5-di-tert-butyl as an electron releasing group. Further, Cu2+ ions play a pivotal role in extracellular aggregation of Aβ42 peptides. So far, we know probably that there are no promising drugs available in this regard. Hence, countering the Cu2+ ions by RHTB chelation against orally administered Cu2+ ion-induced neurotoxicity in the eye tissue of Drosophila expressing human Aβ42 (amyloid-β42) has been tested. The present study involves in vivo and in silico approaches. They reveal the therapeutic potential of RHTB against Cu2+ ion-induced Aβ42 toxicity in Alzheimer's disease (AD) model of Drosophila.
Collapse
Affiliation(s)
- Brijesh Singh Chauhan
- Cancer and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Abhishek Rai
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Avinash Kumar Sonkar
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Kamini Tripathi
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Sonal Upadhyay
- Cancer and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Lallan Mishra
- Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Saripella Srikrishna
- Cancer and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
19
|
Mitra S, Talukdar K, Prasad P, Misra SK, Khan S, Sharp JS, Jurss JW, Chakraborty S. Rational Design of a Cu Chelator That Mitigates Cu-Induced ROS Production by Amyloid Beta. Chembiochem 2022; 23:e202100485. [PMID: 34878720 PMCID: PMC9040527 DOI: 10.1002/cbic.202100485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/07/2021] [Indexed: 11/07/2022]
Abstract
Alzheimer's disease severely perturbs transition metal homeostasis in the brain leading to the accumulation of excess metals in extracellular and intraneuronal locations. The amyloid beta protein binds these transition metals, ultimately causing severe oxidative stress in the brain. Metal chelation therapy is an approach to sequester metals from amyloid beta and relieve the oxidative stress. Here we have designed a mixed N/O donor Cu chelator inspired by the proposed ligand set of Cu in amyloid beta. We demonstrate that the chelator effectively removes Cu from amyloid beta and suppresses reactive oxygen species (ROS) production by redox silencing and radical scavenging both in vitro and in cellulo. The impact of ROS on the extent of oxidation of the different aggregated forms of the peptide is studied by mass spectrometry, which, along with other ROS assays, shows that the oligomers are pro-oxidants in nature. The aliphatic Leu34, which was previously unobserved, has been identified as a new oxidation site.
Collapse
Affiliation(s)
- Suchitra Mitra
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA
| | - Kallol Talukdar
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA
| | - Pallavi Prasad
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA
| | - Sandeep K. Misra
- Department of Biomolecular Sciences, University of Mississippi, University, MS 38677, USA
| | - Shabana Khan
- National Center for Natural Products Research, University of Mississippi, University, MS 38677, USA
| | - Joshua S. Sharp
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA
- Department of Biomolecular Sciences, University of Mississippi, University, MS 38677, USA
| | - Jonah W. Jurss
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA
| | - Saumen Chakraborty
- Department of Chemistry and Biochemistry, University of Mississippi, University, MS 38677, USA
| |
Collapse
|
20
|
Gupta MK, Jena CK, Balachandra C, Sharma NK. Unusual Pseudopeptides: Syntheses and Structural Analyses of Ethylenediprolyl Peptides and Their Metal Complexes with Cu(II) Ion. J Org Chem 2021; 86:16327-16336. [PMID: 34783560 DOI: 10.1021/acs.joc.1c01676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The synthetic unnatural amino acids and their peptides as peptidomimetics have shown remarkable structural and functional properties. In the repertoire of synthetic peptides, pseudopeptides have emerged as attractive small peptidomimetics that are capable of forming the characteristic secondary structures in the solid/solution phase, as in natural peptides. This report describes the synthesis and structural analyses of novel pseudopeptides as ethylenediprolyl (etpro) tetra/hexapeptides, comprising a chiral diaminedicarboxylate scaffold. Their NMR and CD spectral analyses strongly support the formation of the β-turn-type structures in organic solvents (ACN/MeOH). Further, the single-crystal X-ray studies of tetrapseudopeptide confirm the formation of a unique self-assembly structure as β-strand type in the solid state through hydrogen bonding. Importantly, their diamine moiety influences the formation of Cu-complexes with Cu(II) ions. A tetrapseudopeptide monocarboxylate-Cu(II) complex forms the single crystal that is studied by the single-crystal X-ray diffractometer. The crystal structure of the tetrapseudopeptide-Cu(II) complex confirms the formation of the distorted square planar geometry structure, almost like the amyloid β(Aβ)-peptide-Cu(II) complex structural geometry. Hence, these etpro-pseudopeptides are emerging peptidomimatics that form β-turn types of structures and metal complexes mainly with Cu(II) ions. These molecules could be considered for the development of peptide-based catalysts and peptide-based therapeutic drug candidates.
Collapse
Affiliation(s)
- Manish K Gupta
- National Institute of Science Education and Research (NISER)-Bhubaneswar, Jatni Campus, Bhubaneswar 752050, India.,Homi Bhabha National Institute (HBNI), Mumbai 400094, India
| | - Chinmay K Jena
- National Institute of Science Education and Research (NISER)-Bhubaneswar, Jatni Campus, Bhubaneswar 752050, India.,Homi Bhabha National Institute (HBNI), Mumbai 400094, India
| | - Chenikkayala Balachandra
- National Institute of Science Education and Research (NISER)-Bhubaneswar, Jatni Campus, Bhubaneswar 752050, India.,Homi Bhabha National Institute (HBNI), Mumbai 400094, India
| | - Nagendra K Sharma
- National Institute of Science Education and Research (NISER)-Bhubaneswar, Jatni Campus, Bhubaneswar 752050, India.,Homi Bhabha National Institute (HBNI), Mumbai 400094, India
| |
Collapse
|
21
|
Gout J, Meuris F, Desbois A, Dorlet P. In vitro coordination of Fe-protoheme with amyloid β is non-specific and exhibits multiple equilibria. J Inorg Biochem 2021; 227:111664. [PMID: 34955310 DOI: 10.1016/j.jinorgbio.2021.111664] [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: 08/07/2021] [Revised: 10/20/2021] [Accepted: 11/02/2021] [Indexed: 10/19/2022]
Abstract
In addition to copper and zinc, heme is thought to play a role in Alzheimer's disease and its metabolism is strongly affected during the course of this disease. Amyloid β, the peptide associated with Alzheimer's disease, was shown to bind heme in vitro with potential catalytic activity linked to oxidative stress. To date, there is no direct determination of the structure of this complex. In this work, we studied the binding mode of heme to amyloid β in different conditions of pH and redox state by using isotopically labelled peptide in combination with advanced magnetic and vibrational spectroscopic methods. Our results show that the interaction between heme and amyloid β leads to a variety of species in equilibrium. The formation of these species seems to depend on many factors suggesting that the binding site is neither very strong nor highly specific. In addition, our data do not support the currently accepted model where a water molecule is bound to the ferric heme as sixth ligand. They also exclude structural models mimicking a peroxidatic site in the amyloid β-Fe-protoheme complexes.
Collapse
Affiliation(s)
- Jérôme Gout
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Laboratoire Stress Oxydant et Détoxication, Gif-sur-Yvette, France
| | - Floriane Meuris
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Laboratoire Stress Oxydant et Détoxication, Gif-sur-Yvette, France
| | - Alain Desbois
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Laboratoire Stress Oxydant et Détoxication, Gif-sur-Yvette, France.
| | - Pierre Dorlet
- CNRS, Aix-Marseille Université, BIP, IMM, Marseille, France; Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Laboratoire Stress Oxydant et Détoxication, Gif-sur-Yvette, France.
| |
Collapse
|
22
|
Behar AE, Sabater L, Baskin M, Hureau C, Maayan G. A Water-Soluble Peptoid Chelator that Can Remove Cu 2+ from Amyloid-β Peptides and Stop the Formation of Reactive Oxygen Species Associated with Alzheimer's Disease. Angew Chem Int Ed Engl 2021; 60:24588-24597. [PMID: 34510664 DOI: 10.1002/anie.202109758] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/01/2021] [Indexed: 12/25/2022]
Abstract
Cu bound to amyloid-β (Aβ) peptides can act as a catalyst for the formation of reactive oxygen species (ROS), leading to neuropathologic degradation associated with Alzheimer's disease (AD). An excellent therapeutic approach is to use a chelator that can selectively remove Cu from Cu-Aβ. This chelator should compete with Zn2+ ions (Zn) that are present in the synaptic cleft while forming a nontoxic Cu complex. Herein we describe P3, a water-soluble peptidomimetic chelator that selectively removes Cu2+ from Cu-Aβ in the presence of Zn and prevent the formation of ROS even in a reductive environment. We demonstrate, based on extensive spectroscopic analysis, that although P3 extracts Zn from Cu,Zn-Aβ faster than it removes Cu, the formed Zn complexes are kinetic products that further dissociate, while CuP3 is formed as an exclusive stable thermodynamic product. Our unique findings, combined with the bioavailability of peptoids, make P3 an excellent drug candidate in the context of AD.
Collapse
Affiliation(s)
- Anastasia E Behar
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| | - Laurent Sabater
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, 31077, Toulouse, France.,Université de Toulouse, 31077, Toulouse, France
| | - Maria Baskin
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, 31077, Toulouse, France.,Université de Toulouse, 31077, Toulouse, France
| | - Galia Maayan
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, Technion City, 3200008, Haifa, Israel
| |
Collapse
|
23
|
Behar AE, Sabater L, Baskin M, Hureau C, Maayan G. A Water‐Soluble Peptoid Chelator that Can Remove Cu
2+
from Amyloid‐β Peptides and Stop the Formation of Reactive Oxygen Species Associated with Alzheimer's Disease. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202109758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Anastasia E. Behar
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| | - Laurent Sabater
- CNRS LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne 31077 Toulouse France
- Université de Toulouse 31077 Toulouse France
| | - Maria Baskin
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| | - Christelle Hureau
- CNRS LCC (Laboratoire de Chimie de Coordination) 205 route de Narbonne 31077 Toulouse France
- Université de Toulouse 31077 Toulouse France
| | - Galia Maayan
- Schulich Faculty of Chemistry Technion—Israel Institute of Technology Technion City 3200008 Haifa Israel
| |
Collapse
|
24
|
Devonport J, Bodnár N, McGown A, Bukar Maina M, Serpell LC, Kállay C, Spencer J, Kostakis GE. Salpyran: A Cu(II) Selective Chelator with Therapeutic Potential. Inorg Chem 2021; 60:15310-15320. [PMID: 34609139 DOI: 10.1021/acs.inorgchem.1c01912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report the rational design of a tunable Cu(II) chelating scaffold, 2-(((2-((pyridin-2-ylmethyl)amino)ethyl)amino)methyl)phenol, Salpyran (HL). This tetradentate ligand is predicated to have suitable permeation, has an extremely high affinity for Cu compared to clioquinol (pCu7.4 = 10.65 vs 5.91), and exhibits excellent selectivity for Cu(II) over Zn(II) in aqueous media. Solid and solution studies corroborate the formation of a stable [Cu(II)L]+ monocationic species at physiological pH values (7.4). Its action as an antioxidant was tested in ascorbate, tau, and human prion protein assays, which reveal that Salpyran prevents the formation of reactive oxygen species from the binary Cu(II)/H2O2 system, demonstrating its potential use as a therapeutic small molecule metal chelator.
Collapse
Affiliation(s)
- Jack Devonport
- Department of Chemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QJ, United Kingdom
| | - Nikolett Bodnár
- Department of Inorganic and Analytical Chemistry, University of Debrecen, H-4032 Debrecen, Hungary
| | - Andrew McGown
- Department of Chemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QJ, United Kingdom
| | - Mahmoud Bukar Maina
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, United Kingdom.,College of Medical Sciences, Yobe State University, KM 7, Sir Kashim Ibrahim Way, PMB 1144 Damaturu, Yobe State, Nigeria
| | - Louise C Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, United Kingdom
| | - Csilla Kállay
- Department of Inorganic and Analytical Chemistry, University of Debrecen, H-4032 Debrecen, Hungary
| | - John Spencer
- Department of Chemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QJ, United Kingdom
| | - George E Kostakis
- Department of Chemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QJ, United Kingdom
| |
Collapse
|
25
|
Zabiszak M, Frymark J, Nowak M, Grajewski J, Stachowiak K, Kaczmarek MT, Jastrząb R. Influence of d-Electron Divalent Metal Ions in Complex Formation with L-Tartaric and L-Malic Acids. Molecules 2021; 26:5290. [PMID: 34500723 PMCID: PMC8433706 DOI: 10.3390/molecules26175290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/30/2022] Open
Abstract
Binary complexes of α-hydroxy acids (L-Tartaric acid and L-Malic acid) with d-electron metal ions (copper, cobalt, nickel) were investigated. Potentiometric measurements have been performed in aqueous solution with computer analysis of the data for determination of the stability constants of complexes formed in the studied systems. The coordination mode of the complexes was defined using spectroscopic methods: electron paramagnetic resonance (EPR), ultraviolet-visible (UV-Vis), circular dichroism (CD), and infrared (IR). Results of the equilibrium studies have provided evidence for the formation of dimers with copper(II) ions and monomers with cobalt(II) and nickel(II) ions.
Collapse
Affiliation(s)
- Michał Zabiszak
- Faculty of Chemistry, Adami Mickiewicz University in Poznan, Uniwersytetu Poznańskiego 8, 61-614 Poznan, Poland; (J.F.); (M.N.); (J.G.); (K.S.); (M.T.K.); (R.J.)
| | | | | | | | | | | | | |
Collapse
|
26
|
Everett J, Lermyte F, Brooks J, Tjendana-Tjhin V, Plascencia-Villa G, Hands-Portman I, Donnelly JM, Billimoria K, Perry G, Zhu X, Sadler PJ, O'Connor PB, Collingwood JF, Telling ND. Biogenic metallic elements in the human brain? SCIENCE ADVANCES 2021; 7:eabf6707. [PMID: 34108207 PMCID: PMC8189590 DOI: 10.1126/sciadv.abf6707] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/22/2021] [Indexed: 05/12/2023]
Abstract
The chemistry of copper and iron plays a critical role in normal brain function. A variety of enzymes and proteins containing positively charged Cu+, Cu2+, Fe2+, and Fe3+ control key processes, catalyzing oxidative metabolism and neurotransmitter and neuropeptide production. Here, we report the discovery of elemental (zero-oxidation state) metallic Cu0 accompanying ferromagnetic elemental Fe0 in the human brain. These nanoscale biometal deposits were identified within amyloid plaque cores isolated from Alzheimer's disease subjects, using synchrotron x-ray spectromicroscopy. The surfaces of nanodeposits of metallic copper and iron are highly reactive, with distinctly different chemical and magnetic properties from their predominant oxide counterparts. The discovery of metals in their elemental form in the brain raises new questions regarding their generation and their role in neurochemistry, neurobiology, and the etiology of neurodegenerative disease.
Collapse
Affiliation(s)
- James Everett
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Thornburrow Drive, Keele University, Staffordshire ST4 7QB, UK
- School of Engineering, Library Road, University of Warwick, Coventry CV4 7AL, UK
| | - Frederik Lermyte
- School of Engineering, Library Road, University of Warwick, Coventry CV4 7AL, UK
- Department of Chemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, 64287 Darmstadt, Germany
| | - Jake Brooks
- School of Engineering, Library Road, University of Warwick, Coventry CV4 7AL, UK
| | - Vindy Tjendana-Tjhin
- School of Engineering, Library Road, University of Warwick, Coventry CV4 7AL, UK
| | - Germán Plascencia-Villa
- Department of Biology and Neurosciences Institute, The University of Texas at San Antonio (UTSA), San Antonio, TX 78249, USA
| | - Ian Hands-Portman
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry CV4 7AL, UK
| | - Jane M Donnelly
- School of Engineering, Library Road, University of Warwick, Coventry CV4 7AL, UK
| | - Kharmen Billimoria
- School of Engineering, Library Road, University of Warwick, Coventry CV4 7AL, UK
- Department of Chemistry, Library Road, University of Warwick, Coventry CV4 7AL, UK
- LGC Ltd., Queens Road, Teddington TW11 0LY, UK
| | - George Perry
- Department of Biology and Neurosciences Institute, The University of Texas at San Antonio (UTSA), San Antonio, TX 78249, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Peter J Sadler
- Department of Chemistry, Library Road, University of Warwick, Coventry CV4 7AL, UK
| | - Peter B O'Connor
- Department of Chemistry, Library Road, University of Warwick, Coventry CV4 7AL, UK
| | - Joanna F Collingwood
- School of Engineering, Library Road, University of Warwick, Coventry CV4 7AL, UK
| | - Neil D Telling
- School of Pharmacy and Bioengineering, Guy Hilton Research Centre, Thornburrow Drive, Keele University, Staffordshire ST4 7QB, UK.
| |
Collapse
|
27
|
Wang J, Chen X, Bai W, Wang Z, Xiao W, Zhu J. Study on Mechanism of Ginkgo biloba L. Leaves for the Treatment of Neurodegenerative Diseases Based on Network Pharmacology. Neurochem Res 2021; 46:1881-1894. [PMID: 33988813 DOI: 10.1007/s11064-021-03315-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/19/2021] [Accepted: 03/24/2021] [Indexed: 01/20/2023]
Abstract
Ginkgo biloba L. leaves (GBLs), as widely used plant extract sources, significantly improve cognitive, learning and memory function in patients with dementia. However, few studies have been conducted on the specific mechanism of Neurodegenerative diseases (NDs). In this study, network pharmacology was employed to elucidate potential mechanism of GBLs in the treatment of NDs. Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) was used to obtain the chemical components in accordance with the screening principles of oral availability and drug-like property. Potential targets of GBLs were integrated with disease targets, and intersection targets were exactly the potential action targets of GBLs for treating NDs; these key targets were enriched and analyzed by the protein protein interaction (PPI) analysis and molecular docking verification. Key genes were ultimately used to find the biological pathway and explain the therapeutic mechanism by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Twenty-seven active components of GBLs may affect biological processes such as oxidative reactions and activate transcription factor activities. These components may also affect 120 metabolic pathways, such as the PI3K/AKT pathway, by regulating 147 targets, including AKT1, ALB, HSP90AA1, PTGS2, MMP9, EGFR and APP. By using the software iGEMDOCK, the main target proteins were found to bind well to the main active components of GBLs. GBLs have the characteristics of multi-component and multi-target synergistic effect on the treatment of NDs, which preliminarily predicted its possible molecular mechanism of action, and provided the basis for the follow-up study.
Collapse
Affiliation(s)
- Jing Wang
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, Liaoning, People's Republic of China.,Institute of Chemistry and Applications of Plant Resources, Dalian Polytechnic University, Dalian, 116034, Liaoning, People's Republic of China.,Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China.,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China
| | - Xialin Chen
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China.,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China
| | - Weirong Bai
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China.,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China
| | - Zhenzhong Wang
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China.,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang, 222000, Jiangsu, People's Republic of China. .,State Key Laboratory of Pharmaceutical New-tech for Chinese Medicine, Lianyungang, 222000, Jiangsu, People's Republic of China.
| | - Jingbo Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, 116034, Liaoning, People's Republic of China. .,Institute of Chemistry and Applications of Plant Resources, Dalian Polytechnic University, Dalian, 116034, Liaoning, People's Republic of China.
| |
Collapse
|
28
|
Tau/Aβ chimera peptides: A Thioflavin-T and MALDI-TOF study of Aβ amyloidosis in the presence of Cu(II) or Zn(II) ions and total lipid brain extract (TLBE) vesicles. Chem Phys Lipids 2021; 237:105085. [PMID: 33895131 DOI: 10.1016/j.chemphyslip.2021.105085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 02/08/2023]
Abstract
Currently, Alzheimer's Disease (AD) is a complex neurodegenerative condition, with limited therapeutic options. Several factors, like Amyloid β (Aβ) aggregation, tau protein hyperphosphorylation, bio-metals dyshomeostasis and oxidative stress contribute to AD pathogenesis. These pathogenic processes might occur in the aqueous phase but also on neuronal membranes. Thus, investigating the connection between Aβ and biomembranes, becomes important for unveiling the molecular mechanism underlying Aβ amyloidosis as a critical event in AD pathology. In this work, the interaction of two peptides, made up with hybrid sequences from Tau protein 9-16 (EVMEDHAG) or 26-33 (QGGYTMHQ) N-terminal domain and Aβ16-20 (KLVFF) hydrophobic region, with full length Aβ40 or Aβ42 peptides is reported. The studied "chimera" peptides Ac-EVMEDHAGKLVFF-NH2 (τ9-16-KL) and Ac-QGGYTMHQKLVFF-NH2 (τ26-33-KL) are endowed with Aβ recognition and metal ion interaction capabilities provided by the tau or Aβ sequences, respectively. These peptides were characterized in previous study along with their metal dependent interaction and amyloidogenesis, either in the presence or absence of metal ion and artificial membranes made up with Total Lipid Brain Extract (TLBE) components, (Sciacca et al., 2020). In the present paper, the ability of the two peptides to inhibit Aβ aggregation is studied using composite experimental conditions including aqueous solution, the presence of metal ions (Cu or Zn), the presence of lipid vesicles mimicking neuronal membranes as well as the co-presence of metals and TLBE artificial membranes. We used Thioflavine-T (ThT) fluorescence or MALDI-TOF spectrometry analysis of Aβ limited proteolysis to respectively monitor the Aβ aggregation kinetic or validation of the Aβ interacting regions. We demonstrate that τ9-16-KL and τ26-33-KL peptides differently affect Aβ aggregation kinetics, with the tau sequence playing a crucial role. The results are discussed in terms of chimera's peptides hydrophobicity and electrostatic driven interactions at the aqueous/membrane interface.
Collapse
|
29
|
García-Viñuales S, Sciacca MFM, Lanza V, Santoro AM, Grasso G, Tundo GR, Sbardella D, Coletta M, Grasso G, La Rosa C, Milardi D. The interplay between lipid and Aβ amyloid homeostasis in Alzheimer's Disease: risk factors and therapeutic opportunities. Chem Phys Lipids 2021; 236:105072. [PMID: 33675779 DOI: 10.1016/j.chemphyslip.2021.105072] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/15/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022]
Abstract
Alzheimer's Diseases (AD) is characterized by the accumulation of amyloid deposits of Aβ peptide in the brain. Besides genetic background, the presence of other diseases and an unhealthy lifestyle are known risk factors for AD development. Albeit accumulating clinical evidence suggests that an impaired lipid metabolism is related to Aβ deposition, mechanistic insights on the link between amyloid fibril formation/clearance and aberrant lipid interactions are still unavailable. Recently, many studies have described the key role played by membrane bound Aβ assemblies in neurotoxicity. Moreover, it has been suggested that a derangement of the ubiquitin proteasome pathway and autophagy is significantly correlated with toxic Aβ aggregation and dysregulation of lipid levels. Thus, studies focusing on the role played by lipids in Aβ aggregation and proteostasis could represent a promising area of investigation for the design of valuable treatments. In this review we examine current knowledge concerning the effects of lipids in Aβ aggregation and degradation processes, focusing on the therapeutic opportunities that a comprehensive understanding of all biophysical, biochemical, and biological processes involved may disclose.
Collapse
Affiliation(s)
| | - Michele F M Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy
| | - Valeria Lanza
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy
| | - Anna Maria Santoro
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy
| | - Giulia Grasso
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy
| | - Grazia R Tundo
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | - Massimiliano Coletta
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Grasso
- Department of Chemistry, University of Catania, Catania, Italy
| | - Carmelo La Rosa
- Department of Chemistry, University of Catania, Catania, Italy
| | - Danilo Milardi
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania, Italy.
| |
Collapse
|
30
|
Nguyen PH, Ramamoorthy A, Sahoo BR, Zheng J, Faller P, Straub JE, Dominguez L, Shea JE, Dokholyan NV, De Simone A, Ma B, Nussinov R, Najafi S, Ngo ST, Loquet A, Chiricotto M, Ganguly P, McCarty J, Li MS, Hall C, Wang Y, Miller Y, Melchionna S, Habenstein B, Timr S, Chen J, Hnath B, Strodel B, Kayed R, Lesné S, Wei G, Sterpone F, Doig AJ, Derreumaux P. Amyloid Oligomers: A Joint Experimental/Computational Perspective on Alzheimer's Disease, Parkinson's Disease, Type II Diabetes, and Amyotrophic Lateral Sclerosis. Chem Rev 2021; 121:2545-2647. [PMID: 33543942 PMCID: PMC8836097 DOI: 10.1021/acs.chemrev.0c01122] [Citation(s) in RCA: 420] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein misfolding and aggregation is observed in many amyloidogenic diseases affecting either the central nervous system or a variety of peripheral tissues. Structural and dynamic characterization of all species along the pathways from monomers to fibrils is challenging by experimental and computational means because they involve intrinsically disordered proteins in most diseases. Yet understanding how amyloid species become toxic is the challenge in developing a treatment for these diseases. Here we review what computer, in vitro, in vivo, and pharmacological experiments tell us about the accumulation and deposition of the oligomers of the (Aβ, tau), α-synuclein, IAPP, and superoxide dismutase 1 proteins, which have been the mainstream concept underlying Alzheimer's disease (AD), Parkinson's disease (PD), type II diabetes (T2D), and amyotrophic lateral sclerosis (ALS) research, respectively, for many years.
Collapse
Affiliation(s)
- Phuong H Nguyen
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Ayyalusamy Ramamoorthy
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Bikash R Sahoo
- Biophysics and Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109-1055, United States
| | - Jie Zheng
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Peter Faller
- Institut de Chimie, UMR 7177, CNRS-Université de Strasbourg, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | - John E Straub
- Department of Chemistry, Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Laura Dominguez
- Facultad de Química, Departamento de Fisicoquímica, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Joan-Emma Shea
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - Nikolay V Dokholyan
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
- Department of Chemistry, and Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Alfonso De Simone
- Department of Life Sciences, Imperial College London, London SW7 2AZ, U.K
- Molecular Biology, University of Naples Federico II, Naples 80138, Italy
| | - Buyong Ma
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute, Frederick, Maryland 21702, United States
- Sackler Institute of Molecular Medicine, Department of Human Genetics and Molecular Medicine Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Saeed Najafi
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - Son Tung Ngo
- Laboratory of Theoretical and Computational Biophysics & Faculty of Applied Sciences, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
| | - Antoine Loquet
- Institute of Chemistry & Biology of Membranes & Nanoobjects, (UMR5248 CBMN), CNRS, Université Bordeaux, Institut Européen de Chimie et Biologie, 33600 Pessac, France
| | - Mara Chiricotto
- Department of Chemical Engineering and Analytical Science, University of Manchester, Manchester M13 9PL, U.K
| | - Pritam Ganguly
- Department of Chemistry and Biochemistry, and Department of Physics, University of California, Santa Barbara, California 93106, United States
| | - James McCarty
- Chemistry Department, Western Washington University, Bellingham, Washington 98225, United States
| | - Mai Suan Li
- Institute for Computational Science and Technology, SBI Building, Quang Trung Software City, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City 700000, Vietnam
- Institute of Physics, Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Carol Hall
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695-7905, United States
| | - Yiming Wang
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695-7905, United States
| | - Yifat Miller
- Department of Chemistry and The Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Be'er Sheva 84105, Israel
| | | | - Birgit Habenstein
- Institute of Chemistry & Biology of Membranes & Nanoobjects, (UMR5248 CBMN), CNRS, Université Bordeaux, Institut Européen de Chimie et Biologie, 33600 Pessac, France
| | - Stepan Timr
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Jiaxing Chen
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Brianna Hnath
- Department of Pharmacology and Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, and Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Sylvain Lesné
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Guanghong Wei
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Science, Multiscale Research Institute of Complex Systems, Fudan University, Shanghai 200438, China
| | - Fabio Sterpone
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
| | - Andrew J Doig
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, U.K
| | - Philippe Derreumaux
- CNRS, UPR9080, Université de Paris, Laboratory of Theoretical Biochemistry, IBPC, Fondation Edmond de Rothschild, PSL Research University, Paris 75005, France
- Laboratory of Theoretical Chemistry, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, 33000 Ho Chi Minh City, Vietnam
| |
Collapse
|
31
|
Balogh BD, Szakács B, Di Natale G, Tabbì G, Pappalardo G, Sóvágó I, Várnagy K. Copper (II) binding properties of an octapeptide fragment from the R3 region of tau protein: A combined potentiometric, spectroscopic and mass spectrometric study. J Inorg Biochem 2021; 217:111358. [PMID: 33588277 DOI: 10.1016/j.jinorgbio.2021.111358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/10/2021] [Accepted: 01/16/2021] [Indexed: 12/25/2022]
Abstract
The copper(II) complexes of a peptide fragment of the R3 domain of tau protein (tau(326-333) Ac-GNIHHKPG-NH2) and its mutants (Ac-GNGHHKPG-NH2, Ac-GNIHHKAG-NH2, Ac-GNGAHKPG-NH2 and Ac-GNGHAKPG-NH2) have been studied by potentiometric and spectroscopic (UV-Vis, CD) methods. ESR spectroscopy and mass spectrometry were also used to prove the coordination mode of the mononuclear complexes and the formation of dinuclear species, respectively. It has been demonstrated that the (326-333) fragment of tau protein is a versatile and effective ligand for copper(II) coordination. The versatility of copper(II) binding is related to the presence of two adjacent histidyl residues in the sequence, which results in the coexistence of mononuclear, bis(ligand) and dinuclear complexes at different metal to ligand ratios. The 1:1 mononuclear complexes are, however, the dominant species with all peptides and the imidazole-N and one to three deprotonated amide nitrogen atoms towards the N-terminal side of the histidyl residue have been suggested as metal binding sites. This binding mode allows the formation of coordination isomers because any of the two histidine moieties can be the primary anchoring site. It is evident from the CD spectroscopic measurements that the isomers are present in almost equal concentration. The copper(II) binding affinity of the native fragment of tau protein is comparable to that of a similar 2-histidine fragment of amyloid-β mutant, Ac-SGAEGHHQK-NH2 but the comparison with an independent histidyl residue (H32) from the N-terminal region of the protein reveals the predominance of H32 over the histidines in the R3 domain.
Collapse
Affiliation(s)
- Bettina Diána Balogh
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Bence Szakács
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Giuseppe Di Natale
- CNR-Istituto di Cristallografia (IC), s.s. Catania, Via Paolo Gaifami 18, I-95126 Catania, Italy
| | - Giovanni Tabbì
- CNR-Istituto di Cristallografia (IC), s.s. Catania, Via Paolo Gaifami 18, I-95126 Catania, Italy
| | - Giuseppe Pappalardo
- CNR-Istituto di Cristallografia (IC), s.s. Catania, Via Paolo Gaifami 18, I-95126 Catania, Italy
| | - Imre Sóvágó
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Katalin Várnagy
- Department of Inorganic and Analytical Chemistry, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary.
| |
Collapse
|
32
|
Esmieu C, Balderrama-Martínez-Sotomayor R, Conte-Daban A, Iranzo O, Hureau C. Unexpected Trends in Copper Removal from Aβ Peptide: When Less Ligand Is Better and Zn Helps. Inorg Chem 2021; 60:1248-1256. [PMID: 33400522 DOI: 10.1021/acs.inorgchem.0c03407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cu, Zn, and amyloid-β (Aβ) peptides play an important role in the etiology of Alzheimer's disease (AD). Their interaction indeed modifies the self-assembly propensity of the peptide that is at the origin of the deposition of insoluble peptide aggregates in the amyloid plaque, a hallmark found in AD brains. Another even more important fallout of the Cu binding to Aβ peptide is the formation of reactive oxygen species (ROS) that contributes to the overall oxidative stress detected in the disease and is due to the redox ability of the Cu ions. Many therapeutic approaches are currently developed to aid fighting against AD, one of them targeting the redox-active Cu ions. Along this research line, we report in the present article the use of a phenanthroline-based peptide-like ligand (L), which is able to withdraw Cu from Aβ and redox-silence it in a very stable 4N Cu(II) binding site even in the presence of Zn(II). In addition and in contrast to what is usually observed, the presence of excess of L lessens the searched effect of ROS production prevention, but it is counterbalanced by the co-presence of Zn(II). To explain such unprecedented trends, we proposed a mechanism that involves the redox reaction between Cu(II)L and Cu(I)L2. We thus illustrated (i) how speciation and redox chemistry can weaken the effect of a ligand that would have appeared perfectly suitable if only tested in a 1:1 ratio and on CuAβ and (ii) how Zn overcomes the undesired lessening of ROS arrest due to excess of ligand. In brief, we have shown how working in biologically relevant conditions is important for the understanding of all of the reactions at play and this must be taken into consideration for the further rational design of ligands aiming to become drug candidates.
Collapse
Affiliation(s)
- Charlène Esmieu
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077 Toulouse, Cedex 4, France
| | | | - Amandine Conte-Daban
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077 Toulouse, Cedex 4, France
| | - Olga Iranzo
- Aix Marseille University, CNRS, Centrale Marseille, iSm2, Campus Scientifique de St Jérôme, 13397 Marseille, France
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, 31077 Toulouse, Cedex 4, France
| |
Collapse
|
33
|
The Aggregation Pattern of Aβ
1–40
is Altered by the Presence of
N
‐Truncated Aβ
4–40
and/or Cu
II
in a Similar Way through Ionic Interactions. Chemistry 2021; 27:2798-2809. [DOI: 10.1002/chem.202004484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Indexed: 12/19/2022]
|
34
|
Lakey-Beitia J, Burillo AM, Penna GL, Hegde ML, Rao K. Polyphenols as Potential Metal Chelation Compounds Against Alzheimer's Disease. J Alzheimers Dis 2021; 82:S335-S357. [PMID: 32568200 PMCID: PMC7809605 DOI: 10.3233/jad-200185] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease affecting more than 50 million people worldwide. The pathology of this multifactorial disease is primarily characterized by the formation of amyloid-β (Aβ) aggregates; however, other etiological factors including metal dyshomeostasis, specifically copper (Cu), zinc (Zn), and iron (Fe), play critical role in disease progression. Because these transition metal ions are important for cellular function, their imbalance can cause oxidative stress that leads to cellular death and eventual cognitive decay. Importantly, these transition metal ions can interact with the amyloid-β protein precursor (AβPP) and Aβ42 peptide, affecting Aβ aggregation and increasing its neurotoxicity. Considering how metal dyshomeostasis may substantially contribute to AD, this review discusses polyphenols and the underlying chemical principles that may enable them to act as natural chelators. Furthermore, polyphenols have various therapeutic effects, including antioxidant activity, metal chelation, mitochondrial function, and anti-amyloidogenic activity. These combined therapeutic effects of polyphenols make them strong candidates for a moderate chelation-based therapy for AD.
Collapse
Affiliation(s)
- Johant Lakey-Beitia
- Centre for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Clayton, City of Knowledge, Panama
| | - Andrea M. Burillo
- Centre for Biodiversity and Drug Discovery, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Clayton, City of Knowledge, Panama
| | - Giovanni La Penna
- National Research Council, Institute of Chemistry of Organometallic Compounds, Sesto Fiorentino (FI), Italy
| | - Muralidhar L. Hegde
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX, USA
- Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - K.S. Rao
- Centre for Neuroscience, Instituto de Investigaciones Científicas y Servicios de Alta Tecnología (INDICASAT AIP), Clayton, City of Knowledge, Panama
- Zhongke Jianlan Medical Institute, Hangzhou, Republic of China
| |
Collapse
|
35
|
Maiti BK, Govil N, Kundu T, Moura JJ. Designed Metal-ATCUN Derivatives: Redox- and Non-redox-Based Applications Relevant for Chemistry, Biology, and Medicine. iScience 2020; 23:101792. [PMID: 33294799 PMCID: PMC7701195 DOI: 10.1016/j.isci.2020.101792] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
The designed "ATCUN" motif (amino-terminal copper and nickel binding site) is a replica of naturally occurring ATCUN site found in many proteins/peptides, and an attractive platform for multiple applications, which include nucleases, proteases, spectroscopic probes, imaging, and small molecule activation. ATCUN motifs are engineered at periphery by conjugation to recombinant proteins, peptides, fluorophores, or recognition domains through chemically or genetically, fulfilling the needs of various biological relevance and a wide range of practical usages. This chemistry has witnessed significant growth over the last few decades and several interesting ATCUN derivatives have been described. The redox role of the ATCUN moieties is also an important aspect to be considered. The redox potential of designed M-ATCUN derivatives is modulated by judicious choice of amino acid (including stereochemistry, charge, and position) that ultimately leads to the catalytic efficiency. In this context, a wide range of M-ATCUN derivatives have been designed purposefully for various redox- and non-redox-based applications, including spectroscopic probes, target-based catalytic metallodrugs, inhibition of amyloid-β toxicity, and telomere shortening, enzyme inactivation, biomolecules stitching or modification, next-generation antibiotic, and small molecule activation.
Collapse
Affiliation(s)
- Biplab K. Maiti
- National Institute of Technology Sikkim, Ravangla Campus, Barfung Block, Ravangla Sub Division, South Sikkim 737139, India
| | - Nidhi Govil
- National Institute of Technology Sikkim, Ravangla Campus, Barfung Block, Ravangla Sub Division, South Sikkim 737139, India
| | - Taraknath Kundu
- National Institute of Technology Sikkim, Ravangla Campus, Barfung Block, Ravangla Sub Division, South Sikkim 737139, India
| | - José J.G. Moura
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| |
Collapse
|
36
|
Bellia F, Grasso GI, Ahmed IMM, Oliveri V, Vecchio G. Carnoquinolines Target Copper Dyshomeostasis, Aberrant Protein-Protein Interactions, and Oxidative Stress. Chemistry 2020; 26:16690-16705. [PMID: 32627921 DOI: 10.1002/chem.202001591] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 12/20/2022]
Abstract
Metal dysregulation, oxidative stress, protein modification, and aggregation are factors strictly interrelated and associated with neurodegenerative pathologies. As such, all of these aspects represent valid targets to counteract neurodegeneration and, therefore, the development of metal-binding compounds with other properties to combat multifactorial disorders is definitely on the rise. Herein, the synthesis and in-depth analysis of the first hybrids of carnosine and 8-hydroxyquinoline, carnoquinolines (CarHQs), which combine the properties of the dipeptide with those of 8-hydroxyquinoline, are reported. CarHQs and their copper complexes were characterized through several techniques, such as ESI-MS and NMR, UV/Vis, and circular dichroism spectroscopy. CarHQs can modulate self- and copper-induced amyloid-β aggregation. These hybrids combine the antioxidant activity of their parent compounds. Therefore, they can simultaneously scavenge free radicals and reactive carbonyl species, thanks to the phenolic group and imidazole ring. These results indicate that CarHQs are promising multifunctional candidates for neurodegenerative disorders and they are worthy of further studies.
Collapse
Affiliation(s)
- Francesco Bellia
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, P. Gaifami 18, 95126, Catania, Italy
| | - Giuseppa Ida Grasso
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125, Catania, Italy
| | | | - Valentina Oliveri
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125, Catania, Italy
| | - Graziella Vecchio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria 6, 95125, Catania, Italy
| |
Collapse
|
37
|
Esmieu C, Ferrand G, Borghesani V, Hureau C. Impact of N-Truncated Aβ Peptides on Cu- and Cu(Aβ)-Generated ROS: Cu I Matters! Chemistry 2020; 27:1777-1786. [PMID: 33058356 DOI: 10.1002/chem.202003949] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/07/2020] [Indexed: 12/15/2022]
Abstract
In vitro Cu(Aβ1-x )-induced ROS production has been extensively studied. Conversely, the ability of N-truncated isoforms of Aβ to alter the Cu-induced ROS production has been overlooked, even though they are main constituents of amyloid plaques found in the human brain. N-Truncated peptides at the positions 4 and 11 (Aβ4-x and Aβ11-x ) contain an amino-terminal copper and nickel (ATCUN) binding motif (H2 N-Xxx-Zzz-His) that confer them different coordination sites and higher affinities for CuII compared to the Aβ1-x peptide. It has further been proposed that the role of Aβ4-x peptide is to quench CuII toxicity in the brain. However, the role of CuI coordination has not been investigated to date. In contrast to CuII , CuI coordination is expected to be the same for N-truncated and N-intact peptides. Herein, we report in-depth characterizations and ROS production studies of Cu (CuI and CuII ) complexes of the Aβ4-16 and Aβ11-16 N-truncated peptides. Our findings show that the N-truncated peptides do produce ROS when CuI is present in the medium, albeit to a lesser extent than the unmodified counterpart. In addition, when used as competitor ligands (i.e., in the presence of Aβ1-16 ), the N-truncated peptides are not able to fully preclude Cu(Aβ1-16 )-induced ROS production.
Collapse
Affiliation(s)
- Charlène Esmieu
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077, Toulouse Cedex 4, France
| | - Guillaume Ferrand
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077, Toulouse Cedex 4, France.,UPS, INPT, University of Toulouse, 31077, Toulouse Cedex 4, France
| | - Valentina Borghesani
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077, Toulouse Cedex 4, France.,UPS, INPT, University of Toulouse, 31077, Toulouse Cedex 4, France.,current address: School of Chemistry, University of Birmingham, Edgbaston, B15 2TT, UK
| | - Christelle Hureau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099 31077, Toulouse Cedex 4, France.,UPS, INPT, University of Toulouse, 31077, Toulouse Cedex 4, France
| |
Collapse
|
38
|
Merged Tacrine-Based, Multitarget-Directed Acetylcholinesterase Inhibitors 2015-Present: Synthesis and Biological Activity. Int J Mol Sci 2020; 21:ijms21175965. [PMID: 32825138 PMCID: PMC7504404 DOI: 10.3390/ijms21175965] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 11/17/2022] Open
Abstract
Acetylcholinesterase is an important biochemical enzyme in that it controls acetylcholine-mediated neuronal transmission in the central nervous system, contains a unique structure with two binding sites connected by a gorge region, and it has historically been the main pharmacological target for treatment of Alzheimer's disease. Given the large projected increase in Alzheimer's disease cases in the coming decades and its complex, multifactorial nature, new drugs that target multiple aspects of the disease at once are needed. Tacrine, the first acetylcholinesterase inhibitor used clinically but withdrawn due to hepatotoxicity concerns, remains an important starting point in research for the development of multitarget-directed acetylcholinesterase inhibitors. This review highlights tacrine-based, multitarget-directed acetylcholinesterase inhibitors published in the literature since 2015 with a specific focus on merged compounds (i.e., compounds where tacrine and a second pharmacophore show significant overlap in structure). The synthesis of these compounds from readily available starting materials is discussed, along with acetylcholinesterase inhibition data, relative to tacrine, and structure activity relationships. Where applicable, molecular modeling, to elucidate key enzyme-inhibitor interactions, and secondary biological activity is highlighted. Of the numerous compounds identified, there is a subset with promising preliminary screening results, which should inspire further development and future research in this field.
Collapse
|
39
|
The aroylhydrazone INHHQ prevents memory impairment induced by Alzheimer's-linked amyloid-β oligomers in mice. Behav Pharmacol 2020; 31:738-747. [PMID: 32773452 DOI: 10.1097/fbp.0000000000000578] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Converging evidence indicates that neurotoxicity and memory impairment in Alzheimer's disease is induced by brain accumulation of soluble amyloid-β oligomers (AβOs). Physiological metals are poorly distributed and concentrated in the senile plaques typical of Alzheimer's disease, where they may be coordinated to the amyloid-β peptide (Aβ). Indeed, zinc and copper increase Aβ oligomerization and toxicity. Metal-protein attenuating compounds represent a class of agents proposed for Alzheimer's disease treatment, as they reduce abnormal interactions of metal ions with Aβ, inhibit Aβ oligomerization and prevent deleterious redox reactions in the brain. The present work investigates the protective action of an isoniazid-derived aroylhydrazone, INHHQ, on AβO-induced memory impairment. Systemic administration of a single dose of INHHQ (1 mg/kg) prevented both short-term and long-term memory impairment caused by AβOs in mice. In-vitro studies showed that INHHQ prevents Cu(Aβ)-catalyzed production of reactive oxygen species. Although the mechanism of protection by INHHQ is not yet fully understood at a molecular level, the results reported herein certainly point to the value of aroylhydrazones as promising neuroprotective agents in Alzheimer's disease and related disorders.
Collapse
|
40
|
Novel Perspective on Alzheimer's Disease Treatment: Rosmarinic Acid Molecular Interplay with Copper(II) and Amyloid β. Life (Basel) 2020; 10:life10070118. [PMID: 32698429 PMCID: PMC7400086 DOI: 10.3390/life10070118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease is a severe disorder that affects millions of people worldwide. It is a very debilitating disease with no cure at the moment. The necessity of finding an effective treatment is very demanding, and the entire scientific community is putting in a lot of effort to address this issue. The major hallmark of Alzheimer's disease is the presence of toxic aggregated species in the brain, impaired metal homeostasis, and high levels of oxidative stress. Rosmarinic acid is a well-known potent antioxidant molecule, the efficacy of which has been proved both in vitro and in vivo. In this study, we investigated the possible role played by rosmarinic acid as a mediator of the copper(II)-induced neurotoxicity. Several spectroscopic techniques and biological assays were applied to characterize the metal complexes and to evaluate the cytotoxicity and the mutagenicity of rosmarinic acid and its Cu(II) complex. Our data indicate that rosmarinic acid is able to interfere with the interaction between amyloid β and Cu(II) by forming an original ternary association.
Collapse
|
41
|
Atrián-Blasco E, Cerrada E, Faller P, Laguna M, Hureau C. Role of PTA in the prevention of Cu(amyloid-β) induced ROS formation and amyloid-β oligomerisation in the presence of Zn. Metallomics 2020; 11:1154-1161. [PMID: 31098605 DOI: 10.1039/c9mt00011a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Metal-targeting drugs are being widely explored as a possible treatment for Alzheimer's disease, but most of these ligands are developed to coordinate Cu(ii). In a previous communication (E. Atrián-Blasco, E. Cerrada, A. Conte-Daban, D. Testemale, P. Faller, M. Laguna and C. Hureau, Metallomics, 2015, 7, 1229-1232) we showed another strategy where Cu(i) was targeted with the PTA (1,3,5-triaza-7-phosphaadamantane) ligand that is able to target Cu(ii) as well, reduce it and keep it in a safe complexed species. Removal of Cu(ii) from the amyloid-β peptide prevents the stabilization of oligomers and protofibrils and the complexation of Cu(i) also stops the formation of reactive oxygen species. Besides, zinc, which is found in the synaptic cleft at a higher concentration than copper, can hamper the ability of metal-targeting drug candidates, an issue that is still poorly considered and studied. Here we show that PTA fully retains the above described properties even in the presence of zinc, thus fulfilling an additional pre-requisite for its use as a model of Cu(i)-targeting drug candidates in the Alzheimer's disease context.
Collapse
|
42
|
Alghamdi A, Wellbrock T, Birch DJS, Vyshemirsky V, Rolinski OJ. Cu 2+ Effects on Beta-Amyloid Oligomerisation Monitored by the Fluorescence of Intrinsic Tyrosine. Chemphyschem 2019; 20:3181-3185. [PMID: 31539190 DOI: 10.1002/cphc.201900565] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/02/2019] [Indexed: 12/26/2022]
Abstract
A non-invasive intrinsic fluorescence sensing of the early stages of Alzheimer's beta amyloid peptide aggregation in the presence of copper ions is reported. By using time-resolved fluorescence techniques the formation of beta amyloid-copper complexes and the accelerated peptide aggregation are demonstrated. The shifts in the emission spectral peaks indicate that the peptides exhibit different aggregation pathways than in the absence of copper.
Collapse
Affiliation(s)
- Abeer Alghamdi
- Department of Physics, University of Strathclyde, Glasgow, G4 0NG, UK
| | - Thorben Wellbrock
- Department of Physics, University of Strathclyde, Glasgow, G4 0NG, UK
| | - David J S Birch
- Department of Physics, University of Strathclyde, Glasgow, G4 0NG, UK
| | | | - Olaf J Rolinski
- Department of Physics, University of Strathclyde, Glasgow, G4 0NG, UK
| |
Collapse
|
43
|
Kepp KP, Squitti R. Copper imbalance in Alzheimer’s disease: Convergence of the chemistry and the clinic. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
44
|
Stefaniak E, Bal W. Cu II Binding Properties of N-Truncated Aβ Peptides: In Search of Biological Function. Inorg Chem 2019; 58:13561-13577. [PMID: 31304745 DOI: 10.1021/acs.inorgchem.9b01399] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As life expectancy increases, the number of people affected by progressive and irreversible dementia, Alzheimer's Disease (AD), is predicted to grow. No drug designs seem to be working in humans, apparently because the origins of AD have not been identified. Invoking amyloid cascade, metal ions, and ROS production hypothesis of AD, herein we share our point of view on Cu(II) binding properties of Aβ4-x, the most prevalent N-truncated Aβ peptide, currently known as the main constituent of amyloid plaques. The capability of Aβ4-x to rapidly take over copper from previously tested Aβ1-x peptides and form highly stable complexes, redox unreactive and resistant to copper exchange reactions, prompted us to propose physiological roles for these peptides. We discuss the new findings on the reactivity of Cu(II)Aβ4-x with coexisting biomolecules in the context of synaptic cleft; we suggest that the role of Aβ4-x peptides is to quench Cu(II) toxicity in the brain and maintain neurotransmission.
Collapse
Affiliation(s)
- Ewelina Stefaniak
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Pawińskiego 5a , 02-106 Warsaw , Poland
| | - Wojciech Bal
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Pawińskiego 5a , 02-106 Warsaw , Poland
| |
Collapse
|
45
|
Esmieu C, Guettas D, Conte-Daban A, Sabater L, Faller P, Hureau C. Copper-Targeting Approaches in Alzheimer’s Disease: How To Improve the Fallouts Obtained from in Vitro Studies. Inorg Chem 2019; 58:13509-13527. [DOI: 10.1021/acs.inorgchem.9b00995] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
| | | | | | | | - Peter Faller
- LCC−CNRS, Université de Toulouse, CNRS, Toulouse, France
| | | |
Collapse
|
46
|
Khatua P, Mondal S, Bandyopadhyay S. Effects of Metal Ions on Aβ 42 Peptide Conformations from Molecular Simulation Studies. J Chem Inf Model 2019; 59:2879-2893. [PMID: 31095382 DOI: 10.1021/acs.jcim.9b00098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this study, we investigate the conformational characteristics of full-length Aβ42 peptide monomers in the presence of Na+ and Zn2+ metal ions using atomistic molecular dynamics (MD) simulations with an aim to explore the possible driving forces behind enhanced aggregation rates of the peptides in the presence of salts. The calculations reveal that the presence of metal ions shifts the conformational equilibrium more toward the compact ordered Aβ structures. Such compact ordered structures stabilized by distant nonlocal contacts between two crucial hydrophobic segments, hp1 and hp2, primarily through two important hydrophobic aromatic residues, Phe-19 and Phe-20, are expected to trigger the aggregation process at a faster rate by populating and stabilizing the aggregation prone structures. Formation of a significant number of such distant contacts in the presence of Na+ ions has also been found to result in breaking of the N-terminal helix. On the contrary, binding of Zn2+ ion to Aβ peptide is highly specific, which stabilizes the N-terminal helix instead of breaking it. This explains why the aggregation rate of Aβ peptides is higher in the presence of divalent Zn2+ ions than monovalent Na+ ions. Relatively higher overall stability of the most populated Aβ peptide monomers in the presence of Zn2+ ions has been found to be associated with specific Zn2+-Aβ binding and significant free energy gain.
Collapse
Affiliation(s)
- Prabir Khatua
- Molecular Modeling Laboratory, Department of Chemistry , Indian Institute of Technology , Kharagpur 721302 , India
| | - Souvik Mondal
- Molecular Modeling Laboratory, Department of Chemistry , Indian Institute of Technology , Kharagpur 721302 , India
| | - Sanjoy Bandyopadhyay
- Molecular Modeling Laboratory, Department of Chemistry , Indian Institute of Technology , Kharagpur 721302 , India.,Centre for Computational and Data Sciences , Indian Institute of Technology , Kharagpur 721302 , India
| |
Collapse
|
47
|
Ahmadi S, Zhu S, Sharma R, Wilson DJ, Kraatz HB. Interaction of metal ions with tau protein. The case for a metal-mediated tau aggregation. J Inorg Biochem 2019; 194:44-51. [DOI: 10.1016/j.jinorgbio.2019.02.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/05/2019] [Accepted: 02/10/2019] [Indexed: 12/18/2022]
|
48
|
Strodel B, Coskuner-Weber O. Transition Metal Ion Interactions with Disordered Amyloid-β Peptides in the Pathogenesis of Alzheimer's Disease: Insights from Computational Chemistry Studies. J Chem Inf Model 2019; 59:1782-1805. [PMID: 30933519 DOI: 10.1021/acs.jcim.8b00983] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Monomers and oligomers of the amyloid-β peptide aggregate to form the fibrils found in the brains of Alzheimer's disease patients. These monomers and oligomers are largely disordered and can interact with transition metal ions, affecting the mechanism and kinetics of amyloid-β aggregation. Due to the disordered nature of amyloid-β, its rapid aggregation, as well as solvent and paramagnetic effects, experimental studies face challenges in the characterization of transition metal ions bound to amyloid-β monomers and oligomers. The details of the coordination chemistry between transition metals and amyloid-β obtained from experiments remain debated. Furthermore, the impact of transition metal ion binding on the monomeric or oligomeric amyloid-β structures and dynamics are still poorly understood. Computational chemistry studies can serve as an important complement to experimental studies and can provide additional knowledge on the binding between amyloid-β and transition metal ions. Many research groups conducted first-principles calculations, ab initio molecular dynamics simulations, quantum mechanics/classical mechanics simulations, and classical molecular dynamics simulations for studying the interplay between transition metal ions and amyloid-β monomers and oligomers. This review summarizes the current understanding of transition metal interactions with amyloid-β obtained from computational chemistry studies. We also emphasize the current view of the coordination chemistry between transition metal ions and amyloid-β. This information represents an important foundation for future metal ion chelator and drug design studies aiming to combat Alzheimer's disease.
Collapse
Affiliation(s)
- Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry (ICS-6) , Forschungszentrum Jülich GmbH , Jülich 52425 , Germany.,Institute of Theoretical and Computational Chemistry , Heinrich Heine University Düsseldorf , Universitätstrasse 1 , Düsseldorf 40225 , Germany
| | - Orkid Coskuner-Weber
- Molecular Biotechnology , Turkish-German University , Sahinkaya Caddesi, No. 86, Beykoz , Istanbul 34820 , Turkey
| |
Collapse
|
49
|
Rana M, Sharma AK. Cu and Zn interactions with Aβ peptides: consequence of coordination on aggregation and formation of neurotoxic soluble Aβ oligomers. Metallomics 2019; 11:64-84. [DOI: 10.1039/c8mt00203g] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The coordination chemistry of transition metal ions (Fe, Cu, Zn) with the amyloid-β (Aβ) peptides has attracted a lot of attention in recent years due to its repercussions in Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Monika Rana
- Department of Chemistry
- Central University of Rajasthan
- Ajmer 305817
- India
| | - Anuj Kumar Sharma
- Department of Chemistry
- Central University of Rajasthan
- Ajmer 305817
- India
| |
Collapse
|
50
|
Kardos J, Héja L, Simon Á, Jablonkai I, Kovács R, Jemnitz K. Copper signalling: causes and consequences. Cell Commun Signal 2018; 16:71. [PMID: 30348177 PMCID: PMC6198518 DOI: 10.1186/s12964-018-0277-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022] Open
Abstract
Copper-containing enzymes perform fundamental functions by activating dioxygen (O2) and therefore allowing chemical energy-transfer for aerobic metabolism. The copper-dependence of O2 transport, metabolism and production of signalling molecules are supported by molecular systems that regulate and preserve tightly-bound static and weakly-bound dynamic cellular copper pools. Disruption of the reducing intracellular environment, characterized by glutathione shortage and ambient Cu(II) abundance drives oxidative stress and interferes with the bidirectional, copper-dependent communication between neurons and astrocytes, eventually leading to various brain disease forms. A deeper understanding of of the regulatory effects of copper on neuro-glia coupling via polyamine metabolism may reveal novel copper signalling functions and new directions for therapeutic intervention in brain disorders associated with aberrant copper metabolism.
Collapse
Affiliation(s)
- Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - Ágnes Simon
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - István Jablonkai
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - Richard Kovács
- Institute of Neurophysiology, Charité-Universitätsmedizin, Berlin, Germany
| | - Katalin Jemnitz
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| |
Collapse
|