1
|
Meng X, Song Q, Liu Z, Liu X, Wang Y, Liu J. Neurotoxic β-amyloid oligomers cause mitochondrial dysfunction-the trigger for PANoptosis in neurons. Front Aging Neurosci 2024; 16:1400544. [PMID: 38808033 PMCID: PMC11130508 DOI: 10.3389/fnagi.2024.1400544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
As the global population ages, the incidence of elderly patients with dementia, represented by Alzheimer's disease (AD), will continue to increase. Previous studies have suggested that β-amyloid protein (Aβ) deposition is a key factor leading to AD. However, the clinical efficacy of treating AD with anti-Aβ protein antibodies is not satisfactory, suggesting that Aβ amyloidosis may be a pathological change rather than a key factor leading to AD. Identification of the causes of AD and development of corresponding prevention and treatment strategies is an important goal of current research. Following the discovery of soluble oligomeric forms of Aβ (AβO) in 1998, scientists began to focus on the neurotoxicity of AβOs. As an endogenous neurotoxin, the active growth of AβOs can lead to neuronal death, which is believed to occur before plaque formation, suggesting that AβOs are the key factors leading to AD. PANoptosis, a newly proposed concept of cell death that includes known modes of pyroptosis, apoptosis, and necroptosis, is a form of cell death regulated by the PANoptosome complex. Neuronal survival depends on proper mitochondrial function. Under conditions of AβO interference, mitochondrial dysfunction occurs, releasing lethal contents as potential upstream effectors of the PANoptosome. Considering the critical role of neurons in cognitive function and the development of AD as well as the regulatory role of mitochondrial function in neuronal survival, investigation of the potential mechanisms leading to neuronal PANoptosis is crucial. This review describes the disruption of neuronal mitochondrial function by AβOs and elucidates how AβOs may activate neuronal PANoptosis by causing mitochondrial dysfunction during the development of AD, providing guidance for the development of targeted neuronal treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinyu Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
2
|
Kumar N, Khatua P, Sinha SK. Can local heating and molecular crowders disintegrate amyloid aggregates? Chem Sci 2024; 15:6095-6105. [PMID: 38665536 PMCID: PMC11040654 DOI: 10.1039/d4sc00103f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024] Open
Abstract
The present study employs a blend of molecular dynamics simulations and a theoretical model to explore the potential disintegration mechanism of a matured Aβ octamer, aiming to offer a strategy to combat Alzheimer's disease. We investigate local heating and crowding effects on Aβ disintegration by selectively heating key Aβ segments and varying the concentration of sodium dodecyl sulphate (SDS), respectively. Despite initiation of disruption, Aβ aggregates resist complete disintegration during local heating due to rapid thermal energy distribution to the surrounding water. Conversely, although SDS molecules effectively inhibit Aβ aggregation at higher concentration through micelle formation, they fail to completely disintegrate the aggregate due to the exceedingly high energy barrier. To address the sampling challenge posed by the formidable energy barrier, we have performed well-tempered metadynamics simulations. Simulations reveal a multi-step disintegration mechanism for the Aβ octamer, suggesting a probable sequence: octamer → pentamer/hexamer ⇌ tetramer → monomer, with a rate-determining step constituting 45 kJ mol-1 barrier during the octamer to pentamer/hexamer transition. Additionally, we have proposed a novel two-state mean-field model based on Ising spins that offers an insight into the kinetics of the Aβ growth process and external perturbation effects on disintegration. Thus, the current simulation study, coupled with the newly introduced mean-field model, offers an insight into the detailed mechanisms underlying the Aβ aggregation process, guiding potential strategies for effective disintegration of Aβ aggregates.
Collapse
Affiliation(s)
- Naresh Kumar
- Department of Chemistry, Theoretical and Computational Biophysical Chemistry Group, Indian Institute of Technology Ropar Rupnagar Punjab 140001 India +91-01881-232066
| | - Prabir Khatua
- Department of Chemistry, GITAM School of Science, GITAM (Deemed to be University) Bengaluru 562163 India
| | - Sudipta Kumar Sinha
- Department of Chemistry, Theoretical and Computational Biophysical Chemistry Group, Indian Institute of Technology Ropar Rupnagar Punjab 140001 India +91-01881-232066
| |
Collapse
|
3
|
Li X, Zhang Y, Wang Y, Zhang S, Zhang L. Molecular Insights into the Inhibition and Disaggregation Effects of EGCG on Aβ40 and Aβ42 Cofibrillation. J Phys Chem B 2024; 128:1843-1853. [PMID: 38359305 DOI: 10.1021/acs.jpcb.3c07232] [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: 02/17/2024]
Abstract
The misfolding and aggregation of amyloid-β (Aβ) peptides play a pivotal role in the pathogenesis of Alzheimer's disease (AD). Aβ40 and Aβ42, the two primary isoforms of Aβ, can not only self-aggregate into homogeneous aggregates but also coaggregate to form mixed fibrils. Epigallocatechin-3-gallate (EGCG), a prominent tea polyphenol, has shown the capability to prevent the self-aggregation of Aβ40 and Aβ42 peptides and disaggregate their homogeneous fibrils. However, its effects on the cofibrillation of Aβ40 and Aβ42 have not yet been explored. Here, we employed molecular dynamic simulations to investigate the effects of EGCG on the coaggregation of Aβ40 and Aβ42, as well as on their mixed fibril. Our findings indicated that EGCG effectively inhibits the codimerization of Aβ40 and Aβ42 primarily by impeding the interchain interaction between the two isoforms. The key binding sites for EGCG on Aβ40 and Aβ42 are the polar residues and aromatic residues, engaging in hydrogen-bond , π-π, and cation-π interactions with EGCG. Additionally, EGCG disaggregates the Aβ40-Aβ42 mixed fibril by reducing its long-range interaction through similar binding sites and interactions as those between EGCG and Aβ40-Aβ42 heterodimers. Our research reveals the comprehensive inhibition and disaggregation effects of EGCG on the cofibrillation of Aβ isoforms, which provides further support for the development of EGCG as an effective antiaggregation agent for AD.
Collapse
Affiliation(s)
- Xuhua Li
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, China
- State Key Laboratory of Surface Physics, Department of Physics, Fudan University, 2005 Songhu Road, Shanghai 200438, China
| | - Yu Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, China
| | - Yuetian Wang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, China
| | - Shengli Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi'an Jiaotong University, 28 West Xianning Road, Xi'an 710049, China
| |
Collapse
|
4
|
Zhang Y, Borch LA, Fischer NH, Meldal M. Hydrodynamic Control of Alzheimer Aβ Fibrillation with Glucosaminic Acid Containing Click-Cyclized β-Bodies. J Am Chem Soc 2024; 146:2654-2662. [PMID: 38126710 DOI: 10.1021/jacs.3c12118] [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: 12/23/2023]
Abstract
It is well established that the dynamic hydration shell plays a vital role in macromolecular functions such as protein-ligand, protein-protein, protein-DNA, and protein-lipid interactions. Here we investigate how the water modality affects conformational changes, solubility, and motion of fibrillar proteins. The hypothesis is that the introduction of a poly hydroxyl amino acid would increase solvation of the fibril forming peptides, preventing their misfolding and aggregation. For the amyloid β (Aβ) peptide, which is considered to be connected with nervous system diseases, including dementia and cognitive decline in Alzheimer's disease, the formation of β-sheet fibrils always occurs with a conformational change and a decrease in the dynamic hydration shell around Aβ(1-42). We present novel cyclic d-amino acid peptides that effectively inhibit fibrillation through affecting the dynamic hydration shell of Aβ(1-42) in vitro. Using de novo design within the software Molecular Operating Environment (MOE), five different peptides that recognize Alzheimer's fibrils were designed and synthesized. Three of them were cyclic all-d-amino acid peptides incorporating the same polyhydroxy building block derived from d-glucosaminic acid (GA). One peptide was the parent cyclic all d-amino acid inhibitor with no GA incorporated, and another was an all l-amino acid linear fibrillation inhibitor. The GA-containing peptides were found to show significantly improved inhibition of Aβ(1-42) aggregation. The inhibition was dramatically improved by the synergistic application of two GA peptides targeting each end of the growing fibril. The present study may facilitate future developments of intervention strategies for Alzheimer's disease and similar neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuan Zhang
- Center for Evolutionary Chemical Biology, Department of Chemistry, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Line A Borch
- Center for Evolutionary Chemical Biology, Department of Chemistry, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Niklas H Fischer
- Center for Evolutionary Chemical Biology, Department of Chemistry, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Morten Meldal
- Center for Evolutionary Chemical Biology, Department of Chemistry, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
5
|
Kalita K, Phukan SJ, Garai S, Sankaranarayanan K. Polyoxometalates Mediated Amyloid Fibrillation Dynamics and Restoration of Enzyme Activity of Hen Egg White Lysozyme Treated under Cold Atmospheric Pressure Plasma. ACS OMEGA 2024; 9:3423-3429. [PMID: 38284079 PMCID: PMC10809371 DOI: 10.1021/acsomega.3c06921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 01/30/2024]
Abstract
Neurodegenerative disorders are one of the most devastating disorders worldwide. Although a definite mechanistic pathway of neurodegenerative disorders is still not clear, it is almost clear that these diseases are initiated by protein misfolding. Hen Egg White Lysozyme (Lyz) can be converted to highly arranged amyloid fibrils and is therefore considered a good model protein for studying protein aggregation in connection to neurodegeneration. In this study, Lyz has been converted to fibrils using He-air gas fed single jet cold atmospheric plasma (CAP). The reactive oxygen species and the reactive nitrogen species produced by the plasma jet interact with the protein molecules and enhance the fibril formation. We monitored the fibrillation kinetics with the Thioflavin T (ThT) assay and observed that fibrils are formed when the samples are treated for 10 min with He-air gas fed CAP. Further, we studied the role of a special class of inorganic nanomaterials called polyoxometalates (POMs) in the process of the Lyz fibrillation using various biophysical techniques. The Keggin POMs used in this study are phosphomolybdic acid (PMA) and silico molybdic acid (SMA). Keggin POMs bring in structural self-assembly of the protein and disrupt the fibrils as evidenced in the ThT assay and TEM analysis. Molecular docking studies together with electrokinetic potential studies show the interactions between POMs and Lyz dominated via hydrogen bonding and electrostatic interactions. The enzyme activity of Lyz was assessed using the substrate Micrococcus lysodeikticus and after treatment with POMs results showed a significant increase in the activity. This study could pave way for looking into Keggin POMs for possible application in neurodegeneration.
Collapse
Affiliation(s)
- Kaberi Kalita
- Physical
Sciences Division, Institute of Advanced
Study in Science and Technology (An Autonomous Institute Under DST,
Government of India), Vigyan Path, Paschim Boragaon, Garchuk, Guwahati, Assam 781035, India
| | - Shankab J. Phukan
- Department
of Chemistry, Institute of Science, Banaras
Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Somenath Garai
- Department
of Chemistry, Institute of Science, Banaras
Hindu University, Varanasi 221005, Uttar Pradesh, India
| | - Kamatchi Sankaranarayanan
- Physical
Sciences Division, Institute of Advanced
Study in Science and Technology (An Autonomous Institute Under DST,
Government of India), Vigyan Path, Paschim Boragaon, Garchuk, Guwahati, Assam 781035, India
| |
Collapse
|
6
|
Sahraei A, Shamsoddini MJ, Mohammadi F, Hassani L. Interaction of gallium, indium, and vanadyl curcumin complexes with hen egg-white lysozyme (HEWL): Mechanistic aspects and evaluation of antiamyloidogenic activity. Biochem Biophys Res Commun 2024; 691:149307. [PMID: 38011821 DOI: 10.1016/j.bbrc.2023.149307] [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: 08/14/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
Many proteins and peptides can aggregate into amyloid fibrils with high-ordered and cross-β rich structure characteristics. Amyloid deposition is a common feature of neurodegenerative diseases called amyloidosis. Various natural polyphenolic compounds such as curcumin exhibited antiamyloidogenic activities, but less researches were focused on the metal complexes of these compounds. In this study, the inhibitory effects of gallium curcumin (Ga(cur)3), indium curcumin (In(cur)3), and vanadyl curcumin (VO(cur)2) on the amyloid fibrillation of hen egg white lysozyme (HEWL) have been investigated. Moreover, the details of binding interactions of these metal complexes with HEWL have been explored. The results of fluorescence quenching analyses revealed that In(cur)3 and VO(cur)2 have much higher binding affinities than Ga(cur)3 toward HEWL. The interactions of these metal complexes were accompanied by partial conformational changes in the tertiary structure of HEWL. The kinetic curves of the fibrillation process demonstrated that In(cur)3 and VO(cur)2 have higher inhibitory effects than Ga(cur)3 on the amyloid fibrillation of HEWL. The strength of binding to HEWL is completely in accordance with inhibitory activities of these metal complexes of curcumin.
Collapse
Affiliation(s)
- Amin Sahraei
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Blvd., Gava Zang, Zanjan, 45137-66731, Iran
| | - Mohammad Javad Shamsoddini
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Blvd., Gava Zang, Zanjan, 45137-66731, Iran
| | - Fakhrossadat Mohammadi
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Blvd., Gava Zang, Zanjan, 45137-66731, Iran.
| | - Leila Hassani
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Blvd., Gava Zang, Zanjan, 45137-66731, Iran
| |
Collapse
|
7
|
Sahraei A, Ehsanfar A, Mohammadi F. Interaction of gallium, indium and vanadyl diacetylcurcumin complexes with lysozyme: mechanistic aspects and evaluation of antiamyloidogenic activity. ROYAL SOCIETY OPEN SCIENCE 2023; 10:230443. [PMID: 38026032 PMCID: PMC10645073 DOI: 10.1098/rsos.230443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023]
Abstract
Diacetylcurcumin as a derivative of curcumin is a strong nitric oxide (NO) and O2-.anion scavenger. One strategy to improve stability of curcumin and its derivatives is complexation with metal. In this study, the binding interactions of gallium diacetylcurcumin (Ga(DAC)3), indium diacetylcurcumin (In(DAC)3), and vanadyl diacetylcurcumin (VO(DAC)2) with hen egg white lysozyme (HEWL) have been investigated. The results of fluorescence quenching analyses revealed that In(DAC)3 and VO(DAC)2 have higher binding affinities than Ga(DAC)3 towards HEWL. The interactions of these metal complexes were not accompanied by considerable conformational changes in the tertiary structure of HEWL. Furthermore, the inhibitory effects of these complexes on the amyloid fibrillation of HEWL were confirmed by the thioflavin T fluorescence assays. The kinetic curves of the fibrillation process illustrated that VO(DAC)2 has the highest inhibitory activity and In(DAC)3 has a significant delaying effect on the formation of amyloid fibrils of HEWL.
Collapse
Affiliation(s)
- Amin Sahraei
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Boulevard, Gava Zang, Zanjan 45137-66731, Iran
| | - Ahmad Ehsanfar
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Boulevard, Gava Zang, Zanjan 45137-66731, Iran
| | - Fakhrossadat Mohammadi
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), 444 Prof. Sobouti Boulevard, Gava Zang, Zanjan 45137-66731, Iran
| |
Collapse
|
8
|
Waris, Hasnat A, Hasan S, Bano S, Sultana S, Ibhadon AO, Khan MZ. Development of nanozyme based sensors as diagnostic tools in clinic applications: a review. J Mater Chem B 2023; 11:6762-6781. [PMID: 37377089 DOI: 10.1039/d3tb00451a] [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: 06/29/2023]
Abstract
Since 1970, many artificial enzymes that imitate the activity and structure of natural enzymes have been discovered. Nanozymes are a group of nanomaterials with enzyme-mimetic properties capable of catalyzing natural enzyme processes. Nanozymes have attracted great interest in biomedicine due to their excellent stability, rapid reactivity, and affordable cost. The enzyme-mimetic activities of nanozymes may be modulated by numerous parameters, including the oxidative state of metal ions, pH, hydrogen peroxide (H2O2) level, and glutathione (GSH) concentration, indicating the tremendous potential for biological applications. This article delivers a comprehensive overview of the advances in the knowledge of nanozymes and the creation of unique and multifunctional nanozymes, and their biological applications. In addition, a future perspective of employing the as-designed nanozymes in biomedical and diagnostic applications is provided, and we also discuss the barriers and constraints for their further therapeutic use.
Collapse
Affiliation(s)
- Waris
- Environmental Research Laboratory, Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
- Industrial Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, 202002, India
| | - Abul Hasnat
- Environmental Research Laboratory, Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
- Industrial Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, 202002, India
| | - Shumaila Hasan
- Department of Chemistry, Integral University, Lucknow-226026, India
| | - Sayfa Bano
- Environmental Research Laboratory, Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
| | - Saima Sultana
- Environmental Research Laboratory, Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
| | - Alex Omo Ibhadon
- School of Engineering, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - Mohammad Zain Khan
- Environmental Research Laboratory, Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
- Industrial Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, Aligarh Muslim University, Aligarh, 202002, India
| |
Collapse
|
9
|
Jamuna NA, Kamalakshan A, Dandekar BR, Chittilappilly Devassy AM, Mondal J, Mandal S. Mechanistic Insight into the Amyloid Fibrillation Inhibition of Hen Egg White Lysozyme by Three Different Bile Acids. J Phys Chem B 2023; 127:2198-2213. [PMID: 36861956 DOI: 10.1021/acs.jpcb.3c00274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Amyloid aggregation of protein is linked to many neurodegenerative diseases. Identification of small molecules capable of targeting amyloidogenic proteins has gained significant importance. Introduction of hydrophobic and hydrogen bonding interactions through site-specific binding of small molecular ligand to protein can effectively modulate the protein aggregation pathway. Here, we investigate the possible roles of three different bile acids, cholic acid (CA), taurocholic acid (TCA), and lithocholic acid (LCA) with varying hydrophobic and hydrogen bonding properties in inhibiting protein fibrillation. Bile acids are an important class of steroid compounds that are synthesized in the liver from cholesterol. Increasing evidence suggests that altered taurine transport, cholesterol metabolism, and bile acid synthesis have strong implications in Alzheimer's disease. We find that the hydrophilic bile acids, CA and TCA (taurine conjugated form of CA), are substantially more efficient inhibitors of lysozyme fibrillation than the most hydrophobic secondary bile acid LCA. Although LCA binds more strongly with the protein and masks the Trp residues more prominently through hydrophobic interactions, the lesser extent of hydrogen bonding interactions at the active site has made LCA a relatively weaker inhibitor of HEWL aggregation than CA and TCA. The introduction of a greater number of hydrogen bonding channels by CA and TCA with several key amino acid residues which are prone to form oligomers and fibrils has weakened the protein's internal hydrogen bonding capabilities for undergoing amyloid aggregation.
Collapse
Affiliation(s)
- Nidhi Anilkumar Jamuna
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| | - Adithya Kamalakshan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| | | | | | | | - Sarthak Mandal
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| |
Collapse
|
10
|
Roy R, Paul S. Illustrating the Effect of Small Molecules Derived from Natural Resources on Amyloid Peptides. J Phys Chem B 2023; 127:600-615. [PMID: 36638829 DOI: 10.1021/acs.jpcb.2c07607] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The onset of amyloidogenic diseases is associated with the misfolding and aggregation of proteins. Despite extensive research, no effective therapeutics are yet available to treat these chronic degenerative diseases. Targeting the aggregation of disease-specific proteins is regarded as a promising new approach to treat these diseases. In the past few years, rapid progress in this field has been made in vitro, in vivo, and in silico to generate potential drug candidates, ranging from small molecules to polymers to nanoparticles. Small molecular probes, mostly those derived from natural sources, have been of particular interest among amyloid inhibitors. Here, we summarize some of the most important natural small molecular probes which can inhibit the aggregation of Aβ, hIAPP, and α-syn peptides and discuss how their binding efficacy and preference for the peptides vary with their structure and conformation. This provides a comprehensive idea of the crucial factors which should be incorporated into the future design of novel drug candidates useful for the treatment of amyloid diseases.
Collapse
Affiliation(s)
- Rituparna Roy
- Department of Chemistry, Indian Institute of Technology, Guwahati Assam-781039, India
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati Assam-781039, India
| |
Collapse
|
11
|
Wu KY, Doan D, Medrano M, Chang CEA. Modeling structural interconversion in Alzheimers' amyloid beta peptide with classical and intrinsically disordered protein force fields. J Biomol Struct Dyn 2022; 40:10005-10022. [PMID: 34152264 DOI: 10.1080/07391102.2021.1939163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A comprehensive understanding of the aggregation mechanism in amyloid beta 42 (Aβ42) peptide is imperative for developing therapeutic drugs to prevent or treat Alzheimer's disease. Because of the high flexibility and lack of native tertiary structures of Aβ42, molecular dynamics (MD) simulations may help elucidate the peptide's dynamics with atomic details and collectively improve ensembles not seen in experiments. We applied microsecond-timescale MD simulations to investigate the dynamics and conformational changes of Aβ42 by using a newly developed Amber force field (ff14IDPSFF). We compared the ff14IDPSFF and the regular ff14SB force field by examining the conformational changes of two distinct Aβ42 monomers in explicit solvent. Conformational ensembles obtained by simulations depend on the force field and initial structure, Aβ42α-helix or Aβ42β-strand. The ff14IDPSFF sampled a high ratio of disordered structures and diverse β-strand secondary structures; in contrast, ff14SB favored helicity during the Aβ42α-helix simulations. The conformations obtained from Aβ42β-strand simulations maintained a balanced content in the disordered and helical structures when simulated by ff14SB, but the conformers clearly favored disordered and β-sheet structures simulated by ff14IDPSFF. The results obtained with ff14IDPSFF qualitatively reproduced the NMR chemical shifts well. In-depth peptide and cluster analysis revealed some characteristic features that may be linked to early onset of the fibril-like structure. The C-terminal region (mainly M35-V40) featured in-registered anti-parallel β-strand (β-hairpin) conformations with tested systems. Our work should expand the knowledge of force field and structure dependency in MD simulations and reveals the underlying structural mechanism-function relationship in Aβ42 peptides. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kingsley Y Wu
- Department of Chemistry, University of California, Riverside, CA, USA
| | - David Doan
- Department of Chemistry, University of California, Riverside, CA, USA
| | - Marco Medrano
- Department of Chemistry, University of California, Riverside, CA, USA
| | - Chia-En A Chang
- Department of Chemistry, University of California, Riverside, CA, USA
| |
Collapse
|
12
|
Khan AN, Nabi F, Ajmal MR, Ali SM, Almutairi FM, Alalawy AI, Khan RH. Moxifloxacin Disrupts and Attenuates Aβ42 Fibril and Oligomer Formation: Plausibly Repositioning an Antibiotic as Therapeutic against Alzheimer's Disease. ACS Chem Neurosci 2022; 13:2529-2539. [PMID: 35930676 DOI: 10.1021/acschemneuro.2c00371] [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] [Indexed: 01/20/2023] Open
Abstract
The aggregation of Aβ42 is established as a key factor in the development of Alzheimer's disease (AD). Consequently, molecules that inhibit aggregation of peptide may lead to therapies to prevent or control AD. Several studies suggest that oligomeric intermediates present during aggregation may be more cytotoxic than fibrils themselves. In this work, we examine the inhibitory activity of an antibiotic MXF on aggregation (fibrils and oligomers) and disaggregation of Aβ42 using various biophysical and microscopic studies. Computational analysis was done to offer mechanistic insight. The amyloid formation of Aβ42 is suppressed by MXF, as demonstrated by the decrease in both the corresponding ThT fluorescence intensity and other biophysical techniques. The lag phase of amyloid formation doubled from 4.53 to 9.66 h in the presence of MXF. The addition of MXF at the completion of the fibrillation reaction, as monitored by ThT, led to a rapid, concentration dependent, exponential decrease in fluorescence signal that was consistent with loss of fibrils. We used TEM to directly demonstrate that MXF caused fibrils to disassemble. Our docking results show that MXF binds to both monomeric and fibrillar forms of Aβ42 with significant affinities. We also observed breaking of fibrils in the presence of MXF through molecular dynamics simulation. These findings suggest that antibiotic MXF could be a promising lead compound with dual role as fibril/oligomer inhibitor and disaggregase for further development as potential repurposed therapeutic against AD.
Collapse
Affiliation(s)
- Asra Nasir Khan
- Interdisciplinary Biotechnology Unit, AMU, Aligarh 202002, India
| | - Faisal Nabi
- Interdisciplinary Biotechnology Unit, AMU, Aligarh 202002, India
| | - Mohammad Rehan Ajmal
- Physical Biochemistry Research Laboratory, Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Syed Moasfar Ali
- Interdisciplinary Biotechnology Unit, AMU, Aligarh 202002, India
| | - Fahad M Almutairi
- Physical Biochemistry Research Laboratory, Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Adel I Alalawy
- Physical Biochemistry Research Laboratory, Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| | | |
Collapse
|
13
|
Nucara A, Ripanti F, Sennato S, Nisini G, De Santis E, Sefat M, Carbonaro M, Mango D, Minicozzi V, Carbone M. Influence of Cortisol on the Fibril Formation Kinetics of Aβ42 Peptide: A Multi-Technical Approach. Int J Mol Sci 2022; 23:ijms23116007. [PMID: 35682687 PMCID: PMC9180743 DOI: 10.3390/ijms23116007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 11/24/2022] Open
Abstract
Amyloid-β peptide (Aβ) aggregates are known to be correlated with pathological neurodegenerative diseases. The fibril formation process of such peptides in solution is influenced by several factors, such as the ionic strength of the buffer, concentration, pH, and presence of other molecules, just to mention a few. In this paper, we report a detailed analysis of in vitro Aβ42 fibril formation in the presence of cortisol at different relative concentrations. The thioflavin T fluorescence assay allowed us to monitor the fibril formation kinetics, while a morphological characterization of the aggregates was obtained by atomic force microscopy. Moreover, infrared absorption spectroscopy was exploited to investigate the secondary structure changes along the fibril formation path. Molecular dynamics calculations allowed us to understand the intermolecular interactions with cortisol. The combined results demonstrated the influence of cortisol on the fibril formation process: indeed, at cortisol-Aβ42 concentration ratio (ρ) close to 0.1 a faster organization of Aβ42 fragments into fibrils is promoted, while for ρ = 1 the formation of fibrils is completely inhibited.
Collapse
Affiliation(s)
- Alessandro Nucara
- Department of Physics, Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy;
- Correspondence: (A.N.); (F.R.)
| | - Francesca Ripanti
- Department of Physics and Geology, University of Perugia, Via Alessandro Pascoli, 06123 Perugia, Italy
- Correspondence: (A.N.); (F.R.)
| | - Simona Sennato
- CNR-ISC Sede Sapienza, Department of Physics, Sapienza University, P.le A. Moro 5, 00185 Rome, Italy;
| | - Giacomo Nisini
- Department of Physics, Sapienza University of Rome, P. le A. Moro 5, 00185 Rome, Italy;
| | - Emiliano De Santis
- Department of Physics and Astronomy and Department of Chemistry-BMC, Uppsala University, Husargatan 3, 752 37 Uppsala, Sweden;
| | - Mahta Sefat
- School of Pharmacy, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (M.S.); (D.M.)
| | - Marina Carbonaro
- Council for Agricultural Research and Economics (CREA), Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy;
| | - Dalila Mango
- School of Pharmacy, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (M.S.); (D.M.)
- Laboratory Pharmacology of Synaptic Plasticity, European Brain Research Institute, 00161 Rome, Italy
| | - Velia Minicozzi
- Department of Physics and INFN, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133 Rome, Italy;
| | - Marilena Carbone
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy;
| |
Collapse
|
14
|
Guo J, Xu A, Cheng M, Wan Y, Wang R, Fang Y, Jin Y, Xie SS, Liu J. Design, Synthesis and Biological Evaluation of New 3,4-Dihydro-2(1H)-Quinolinone-Dithiocarbamate Derivatives as Multifunctional Agents for the Treatment of Alzheimer’s Disease. Drug Des Devel Ther 2022; 16:1495-1514. [PMID: 35611357 PMCID: PMC9124477 DOI: 10.2147/dddt.s354879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/10/2022] [Indexed: 12/01/2022] Open
Abstract
Background Alzheimer’s disease (AD) belongs to neurodegenerative disease, and the increasing number of AD patients has placed a heavy burden on society, which needs to be addressed urgently. ChEs/MAOs dual-target inhibitor has potential to treat AD according to reports. Purpose To obtain effective multi-targeted agents for the treatment of AD, a novel series of hybrid compounds were designed and synthesized by fusing the pharmacophoric features of 3,4-dihydro-2 (1H)-quinolinone and dithiocarbamate. Methods All compounds were evaluated for their inhibitory abilities of ChEs and MAOs. Then, further biological activities of the most promising candidate 3e were determined, including the ability to cross the blood-brain barrier (BBB), kinetics and molecular model analysis, cytotoxicity in vitro and acute toxicity studies in vivo. Results Most compounds showed potent and clear inhibition to AChE and MAOs. Among them, compound 3e was considered to be the most effective and balanced inhibitor to both AChE and MAOs (IC50=0.28 µM to eeAChE; IC50=0.34 µM to hAChE; IC50=2.81 µM to hMAO-B; IC50=0.91 µM to hMAO-A). In addition, 3e showed mixed inhibition of hAChE and competitive inhibition of hMAO-B in the enzyme kinetic studies. Further studies indicated that 3e could penetrate the BBB and showed no toxicity on PC12 cells and HT-22 cells when the concentration of 3e was lower than 12.5 µM. More importantly, 3e lacked acute toxicity in mice even at high dose (2500 mg/kg, P.O.). Conclusion This work indicated that compound 3e with a six-carbon atom linker and a piperidine moiety at terminal position was a promising candidate and was worthy of further study.
Collapse
Affiliation(s)
- Jie Guo
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Airen Xu
- Clinical Pharmacology Research Center, The Second Hospital of Yinzhou, Ningbo, Zhejiang, People’s Republic of China
| | - Maojun Cheng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Yang Wan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Rikang Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Yuanying Fang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Sai-Sai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
- Correspondence: Sai-Sai Xie, National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, No. 56, Yangming Road, Donghu District, Nanchang City, Jiangxi Province, 330006, People’s Republic of China, Email
| | - Jing Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
- Jing Liu, School of Pharmacy, Jiangxi University of Chinese Medicine, No. 56, Yangming Road, Donghu District, Nanchang City, Jiangxi Province, 330006, People’s Republic of China, Email
| |
Collapse
|
15
|
Insights into the cross-amyloid aggregation of Aβ40 and its N-terminal truncated peptide Aβ11-40 affected by epigallocatechin gallate. Chin J Chem Eng 2022. [DOI: 10.1016/j.cjche.2021.04.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
16
|
Mishra P, Basak S, Mukherjee A, Basu A. Design and Study of In Silico Binding Dynamics of Certain Isoxazole Bearing Leads Against Aβ-42 and BACE-1 Loop in Protein Fibrillation. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666210813120444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aims:
Design isoxazole bearing leads as dual inhibitors against Amyloid β and BACE-1 loop
in protein fibrillation.
Background:
Protein fibrillation is one of the key reasons for several diseases, namely Alzheimer’s, Parkinson’s,
and many others. One of the key strategies of preventing protein fibrillation is destabilizing the
protein fibrils themselves or inhibiting the amyloid fibril-forming pathway in the initial stage.
Introduction:
Attempts have been taken to design newer leads to inhibit protein fibrillation by targeting
the β-amyloidogenesis pathway in the brain. To exploit interfenestration between Amyloid β -42 protein
and BACE-1 (β-site amyloid precursor protein cleaving enzyme) for amyloidogenesis, studies are undertaken
to design dual inhibitors against the same.
Method:
In vitro binding interactions were found using docking, de novo ligand design, and MD simulation
study.
Results:
Three compounds bearing an isoxazole heterocyclic nucleus were designed which could successfully
bind to the hydrophobic raft and salt bridge residues Asp 23-Lys-26 of Amyloid β, destabilizing the
growing fibril. Additionally, one of our candidate compounds exhibited force of interaction with Thr232
at the S3 pocket of BACE-1, interacted with key residue Asp228, Tyr71, and Thr72 of the β-hairpin flap
and hydrogen bonding with Gly11 at loop 10s.
Conclusion:
Protein flexibility dynamics of the Aβ-42 protein revealed that there is a considerable conformational
change of the same with or without ligand binding. The lower RMSF of the bound region and
reprogramming residual contacts within the Aβ-42 protein suggested successful binding of the ligand with
the protein, lowering the access for further β-β dimerization.
Collapse
Affiliation(s)
- Puja Mishra
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, WB, India
| | - Souvik Basak
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, WB, India
| | - Arup Mukherjee
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Kolkata, WB, India
| | - Anindya Basu
- School of Pharmaceutical Sciences, Rajiv
Gandhi Proudyogiki Vishwavidyalaya, Bhopal, India
| |
Collapse
|
17
|
Hu S, Yang C, Li Y, Luo Q, Luo H. Nanozyme sensor array based on manganese dioxide for the distinction between multiple amyloid β peptides and their dynamic aggregation process. Biosens Bioelectron 2021; 199:113881. [PMID: 34915216 DOI: 10.1016/j.bios.2021.113881] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/07/2021] [Accepted: 12/09/2021] [Indexed: 01/01/2023]
Abstract
The determination of the amyloid β (Aβ) peptide and its aggregation intermediates helps to understand the pathological mechanism of Alzheimer's disease (AD) caused by toxic amyloid fragments. Because of the transient and heterogeneous properties of Aβ aggregates, it is very difficult to dynamically detect Aβ and its aggregation intermediates. Herein, we successfully constructed a two-dimensional manganese dioxide (MnO2) nanozyme sensor array by modulating the peroxidase-mimicking activity using various Aβ species and accurately distinguished among six types of Aβ within 1 h through linear discriminant analysis (LDA), with a dynamic detection range of 0.01-500 nmol/L and a detection limit of 0.44 pmol/L. Subsequently, 30 unknown blind samples were used to verify the practicability of the sensor array, and all unknown samples were identified with 100% accuracy. It is worth noting that the sensor array successfully distinguished healthy individuals from AD patients using clinical blood samples. This study provides a convenient and reliable nanozyme biosensing system for detecting Aβ species and their related aggregation processes.
Collapse
Affiliation(s)
- Shun Hu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China; MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Changwen Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China; MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqing Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China; MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China; MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China; School of Biomedical Engineering, Hainan University, Haikou, Hainan, 570228, China
| | - Haiming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China; MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
18
|
Fan Q, Gao Y, Mazur F, Chandrawati R. Nanoparticle-based colorimetric sensors to detect neurodegenerative disease biomarkers. Biomater Sci 2021; 9:6983-7007. [PMID: 34528639 DOI: 10.1039/d1bm01226f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neurodegenerative disorders (NDDs) are progressive, incurable health conditions that primarily affect brain cells, and result in loss of brain mass and impaired function. Current sensing technologies for NDD detection are limited by high cost, long sample preparation, and/or require skilled personnel. To overcome these limitations, optical sensors, specifically colorimetric sensors, have garnered increasing attention towards the development of a cost-effective, simple, and rapid alternative approach. In this review, we evaluate colorimetric sensing strategies of NDD biomarkers (e.g. proteins, neurotransmitters, bio-thiols, and sulfide), address the limitations and challenges of optical sensor technologies, and provide our outlook on the future of this field.
Collapse
Affiliation(s)
- Qingqing Fan
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Yuan Gao
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Federico Mazur
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| | - Rona Chandrawati
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| |
Collapse
|
19
|
Li F, Zhan C, Dong X, Wei G. Molecular mechanisms of resveratrol and EGCG in the inhibition of Aβ 42 aggregation and disruption of Aβ 42 protofibril: similarities and differences. Phys Chem Chem Phys 2021; 23:18843-18854. [PMID: 34612422 DOI: 10.1039/d1cp01913a] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The aggregation of amyloid-β protein (Aβ) into fibrillary deposits is implicated in Alzheimer's disease (AD), and inhibiting Aβ aggregation and clearing Aβ fibrils are considered as promising strategies to treat AD. It has been reported that resveratrol (RSV) and epigallocatechin-3-gallate (EGCG), two of the most extensively studied natural polyphenols, are able to inhibit Aβ fibrillization and remodel the preformed fibrillary aggregates into amorphous, non-toxic species. However, the mechanisms by which RSV inhibits Aβ42 aggregation and disrupts Aβ42 protofibril, as well as the inhibitory/disruptive mechanistic similarities and differences between RSV and EGCG, remain mostly elusive. Herein, we performed extensive all-atom molecular dynamics (MD) simulations on Aβ42 dimers (the early aggregation state of Aβ42) and protofibrils (the intermediate of Aβ42 fibril formation and elongation) in the absence/presence of RSV or EGCG molecules. Our simulations show that both RSV and EGCG can bind with Aβ42 monomers and inhibit the dimerization of Aβ42. The binding of RSV with Aβ42 peptide is mostly viaπ-π stacking interactions, while the binding of EGCG with Aβ42 is mainly through hydrophobic, π-π stacking, and hydrogen-bonding interactions. Moreover, both RSV and EGCG disrupt the β-sheet structure and K28-A42 salt bridges, leading to a disruption of Aβ42 protofibril structure. RSV mainly binds with residues whose side-chains point inwards from the surface of the protofibril, while EGCG mostly binds with residues whose side-chains point outwards from the surface of the protofibril. Furthermore, RSV interacts with Aβ42 protofibrils mostly viaπ-π stacking interactions, while EGCG interacts with Aβ42 protofibrils mainly via hydrogen-bonding and hydrophobic interactions. For comparison, we also explore the effects of RSV/EGCG molecules on the aggregation inhibition and protofibril disruption of the Iowa mutant (D23N) Aβ. Our findings may pave the way for the design of more effective drug candidates as well as the utilization of cocktail therapy using RSV and EGCG for the treatment of AD.
Collapse
Affiliation(s)
- Fangying Li
- Department of Physics, State Key Laboratory of Surface Physics, and Key Laboratory for Computational Physical Sciences (Ministry of Education), Fudan University, Shanghai, 200438, People's Republic of China.
| | | | | | | |
Collapse
|
20
|
Design, synthesis and in-vitro evaluation of fluorinated triazoles as multi-target directed ligands for Alzheimer disease. Bioorg Med Chem Lett 2021; 42:127999. [PMID: 33839248 DOI: 10.1016/j.bmcl.2021.127999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 12/31/2022]
Abstract
Alzheimer disease is multi-factorial and inflammation plays a major role in the disease progression and severity. Metals and reactive oxygen species (ROS) are the key mediators for inflammatory conditions associated with Alzheimer's. Along multi-factorial nature, major challenge for developing new drug is the ability of the molecule to cross blood brain barrier (BBB). We have designed and synthesized multi-target directed hexafluorocarbinol containing triazoles to inhibit Amyloid β aggregation and simultaneously chelate the excess metals present in the extracellular space and scavenge the ROS thus reduce the inflammatory condition. From the screened compound library, compound 1c found to be potent and safe. It has demonstrated inhibition of Amyloid β aggregation (IC50 of 4.6 μM) through selective binding with Amyloid β at the nucleation site (evidenced from the molecular docking). It also chelate metals (Cu+2, Zn+2 and Fe+3) and scavenges ROS significantly. Due to the presence of hexafluorocarbinol moiety in the molecule it may assist to permeate BBB and improve the pharmacokinetic properties. The in-vitro results of compound 1c indicate the promiscuity for the development of hexafluorocarbinol containing triazoles amide scaffold as multi-target directed therapy against Alzheimer disease.
Collapse
|
21
|
Li G, Zhou Y, Yang WY, Zhang C, Hong L, Jia L. Inhibitory Effects of Sulfated Polysaccharides from the Sea Cucumber Cucumaria Frondosa against Aβ40 Aggregation and Cytotoxicity. ACS Chem Neurosci 2021; 12:1854-1859. [PMID: 33999600 DOI: 10.1021/acschemneuro.1c00223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Abnormal aggregation and deposition of Aβ is one of the causative agents for Alzheimer's disease. The development of inhibitors for Aβ aggregation has been considered a possible method to prevent and treat Alzheimer's disease. Edible sea cucumbers contain many bioactive molecules, including saponins, phospholipids, peptides, and polysaccharides. Herein, we report that polysaccharides extracted from sea cucumber Cucumaria frondosa could reduce the aggregation and cytotoxicity of Aβ40. By utilizing multiple biochemical and biophysical instruments, we found that the polysaccharides could inhibit the aggregation of Aβ40. A chemical kinetics analysis further suggested that the major inhibitory effects of the polysaccharides were achieved by disassembling mature fibrils, which in turn reduced the cytotoxicity of Aβ. These results suggested that the polysaccharides extracted from sea cucumber could be used as an effective inhibitor for Aβ.
Collapse
Affiliation(s)
- Gao Li
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| | - Yu Zhou
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| | - Wu-Yue Yang
- Yau Mathematical Sciences Center, Tsinghua University, Beijing, Beijing 100084, China
| | - Chen Zhang
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| | - Liu Hong
- School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Lee Jia
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| |
Collapse
|
22
|
Illes-Toth E, Meisl G, Rempel DL, Knowles TPJ, Gross ML. Pulsed Hydrogen-Deuterium Exchange Reveals Altered Structures and Mechanisms in the Aggregation of Familial Alzheimer's Disease Mutants. ACS Chem Neurosci 2021; 12:1972-1982. [PMID: 33988976 DOI: 10.1021/acschemneuro.1c00072] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Mutations of the Amyloid Precursor Protein, from which the amyloid β peptide Aβ42 is cleaved, are associated with familial Alzheimer's disease. The disease-relevant familial mutations include the Arctic (E22G), Iowa (D23N), Italian (E22K), Dutch (E22Q), Japanese (D7N), English (D6R), and Flemish (A21G) variants. A detailed mechanistic understanding of the aggregation behavior of the mutant peptides at the residue level is, however, still lacking. We report here a study of the aggregation kinetics of these mutants in vitro by pulsed hydrogen-deuterium exchange mass spectrometry (HDX-MS) to obtain a temporally and sequence resolved picture of their self-assembly. For all variants, HDX occurs to give a bimodal distribution representing two soluble classes of aggregates, one protected and one solvent-exposed. There is no evidence of other classes of structural intermediates within the detection limits of the HDX approach. The fractional changes in the bimodal exchange profiles for several regions of Aβ42 reveal that the central and C-terminal peptides gain protection upon fibril formation, whereas the N-terminal regions remain largely solvent-accessible. For these mutants, all peptide fragments follow the same kinetics, acquiring solvent protection at the same time, further supporting that there are no significant populations of intermediate species under our experimental conditions. The results demonstrate the potential of pulsed HDX-MS for resolving the region-specific aggregation behavior of Aβ42 isoforms in solution where X-ray crystallography and solid-state NMR (ssNMR) are challenged.
Collapse
Affiliation(s)
- Eva Illes-Toth
- Washington University in St. Louis, Department of Chemistry, St. Louis, Missouri 63130, United States
| | - Georg Meisl
- University of Cambridge, Department of Chemistry, Lensfield Road, Cambridge CB2 1EW, UK
| | - Don L. Rempel
- Washington University in St. Louis, Department of Chemistry, St. Louis, Missouri 63130, United States
| | - Tuomas P. J. Knowles
- University of Cambridge, Department of Chemistry, Lensfield Road, Cambridge CB2 1EW, UK
| | - Michael L. Gross
- Washington University in St. Louis, Department of Chemistry, St. Louis, Missouri 63130, United States
| |
Collapse
|
23
|
Wang W, Zhao G, Dong X, Sun Y. Unexpected Function of a Heptapeptide-Conjugated Zwitterionic Polymer that Coassembles into β-Amyloid Fibrils and Eliminates the Amyloid Cytotoxicity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:18089-18099. [PMID: 33829756 DOI: 10.1021/acsami.1c01132] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Fibrillogenesis of amyloid β-protein (Aβ) is pathologically associated with Alzheimer's disease (AD), so modulating Aβ aggregation is crucial for AD prevention and treatment. Herein, a zwitterionic polymer with short dimethyl side chains (pID) is synthesized and conjugated with a heptapeptide inhibitor (Ac-LVFFARK-NH2, LK7) to construct zwitterionic polymer-inhibitor conjugates for enhanced inhibition of Aβ aggregation. However, it is unexpectedly found that the LK7@pID conjugates remarkably promote Aβ fibrillization to form more fibrils than the free Aβ system but effectively eliminate Aβ-induced cytotoxicity. Such an unusual behavior of the LK7@pID conjugates is unraveled by extensive mechanistic studies. First, the hydrophobic environment within the assembled micelles of LK7@pID promotes the hydrophobic interaction between Aβ molecules and LK7@pID, which triggers Aβ aggregation at the very beginning, making fibrillization occur at an earlier stage. Second, in the aggregation process, the LK7@pID micelles disassemble by the intensive interactions with Aβ, and LK7@pID participates in the fibrillization by being embedded in the Aβ fibrils, leading to the formation of hybrid and heterogeneous fibrillar aggregates with a different structure than normal Aβ fibrils. This unique Trojan horse-like feature of LK7@pID conjugates has not been observed for any other inhibitors reported previously and may shed light on the design of new modulators against β-amyloid cytotoxicity.
Collapse
Affiliation(s)
- Wenjuan Wang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Guangfu Zhao
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| |
Collapse
|
24
|
Kumari A, Shrivastava N, Mishra M, Somvanshi P, Grover A. Inhibitory mechanism of an antifungal drug, caspofungin against amyloid β peptide aggregation: Repurposing via neuroinformatics and an experimental approach. Mol Cell Neurosci 2021; 112:103612. [PMID: 33722677 DOI: 10.1016/j.mcn.2021.103612] [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: 11/15/2020] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 12/01/2022] Open
Abstract
The multifactorial neurological condition called Alzheimer's disease (AD) primarily affects elderly individuals. Despite the calamitous consequences of AD, curative strategies for a regimen to apply remain inadequate as several factors contribute to AD etiology. Drug repurposing is an advance strategy prior to drug discovery as various effective drugs perform through alteration of multiple targets, and the present "poly-pharmacology" can be a curative approach to complex disorders. AD's multifactorial behavior actively encourages the hypothesis for a drug design approach focused on drug repurposing. In this study, we discovered that an antifungal drug, Caspofungin (CAS) is a potent Aβ aggregation inhibitor that displays significantly reduced toxicity associated with AD. Drug reprofiling and REMD simulations demonstrated that CAS interacts with the β-sheet section, known as Aβ amyloid fibrils hotspot. CAS leads to destabilization of β-sheet and, conclusively, in its devaluation. Later, in vitro experiments were acquired in which the fibrillar volume was reduced for CAS-treated Aβ peptide. For the first time ever, this study has determined an antifungal agent as the Aβ amyloid aggregation's potent inhibitor. Several efficient sequence-reliant potent inhibitors can be developed in future against the amyloid aggregation for different amyloid peptide by the processing and conformational optimization of CAS.
Collapse
Affiliation(s)
- Anchala Kumari
- Department of Biotechnology, Teri School of Advanced Studies, New Delhi 110070, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nidhi Shrivastava
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Mohit Mishra
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Pallavi Somvanshi
- Department of Biotechnology, Teri School of Advanced Studies, New Delhi 110070, India.
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
25
|
Liu W, Dong X, Liu Y, Sun Y. Photoresponsive materials for intensified modulation of Alzheimer's amyloid-β protein aggregation: A review. Acta Biomater 2021; 123:93-109. [PMID: 33465508 DOI: 10.1016/j.actbio.2021.01.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/29/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022]
Abstract
The abnormal self-assembly of amyloid-β protein (Aβ) into toxic aggregates is a major pathological hallmark of Alzheimer's disease (AD). Modulation of Aβ fibrillization with pharmacological modalities has become an active field of research, which aims to mitigate Aβ-induced neurotoxicity and ameliorate impaired recognition. Among the various strategies for AD treatment, phototherapy, including photothermal therapy (PTT), photodynamic therapy (PDT), and photoresponsive release systems have attracted increased attention because of the spatiotemporal controllability. Under the irradiation of light, the heat or reactive oxygen species generated by photothermal or photodynamic processes significantly enhances the efficacy of the inhibitor or modulator, and the "caged" drug can be accurately released at the intended site, thus avoiding adverse effects. This review, from a viewpoint of materials, focuses on the recent advances in modulating Aβ aggregation by light that irradiates on the materials that function on modulating Aβ aggregation. Representative examples of PTT, PDT, and photoresponsive drug release systems are discussed in terms of inhibitory mechanism, the unique properties of materials, and the design of modulators. The major challenges of phototherapy against AD are addressed and the promising prospects are proposed. It is concluded that the noninvasive light-assisted approaches will become a promising strategy for intensifying the modulation of Aβ aggregation and thus facilitating AD treatment. STATEMENT OF SIGNIFICANCE: Alzheimer's disease (AD) with the hallmark of amyloid-β protein (Aβ) deposition is affecting more than 50 million people globally. It is urgent to explore intelligent materials to modulate Aβ aggregation. This review summarizes the intensified modulation of Aβ aggregation by a variety of photoresponsive materials including photothermal, photosensitizing and photoresponsive release materials, focusing on their characteristics and functionalities. We believe this review would arouse more interest in the research field of stimuli-responsive materials and promote their clinical applications in AD therapy.
Collapse
Affiliation(s)
- Wei Liu
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Yang Liu
- Department of Biology & Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, College of Science, Shantou University, Shantou, Guangdong 515063, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology and Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China.
| |
Collapse
|
26
|
Wang X, Wang C, Chan HN, Ashok I, Krishnamoorthi SK, Li M, Li HW, Wong MS. Amyloid-β oligomer targeted theranostic probes for in vivo NIR imaging and inhibition of self-aggregation and amyloid-β induced ROS generation. Talanta 2021; 224:121830. [DOI: 10.1016/j.talanta.2020.121830] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022]
|
27
|
Chromone derivatives bearing pyridinium moiety as multi-target-directed ligands against Alzheimer's disease. Bioorg Chem 2021; 110:104750. [PMID: 33691251 DOI: 10.1016/j.bioorg.2021.104750] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/16/2021] [Accepted: 02/13/2021] [Indexed: 01/04/2023]
Abstract
A new serise of 7-hydroxy-chromone derivatives bearing pyridine moiety were synthesized, and evaluated as multifunctional agents against Alzheimer's disease (AD). Most of the compounds were good AChE inhibitors (IC50 = 9.8-0.71 µM) and showed remarkable BuChE inhibition activity (IC50 = 1.9-0.006 µM) compared with donepezil as the standard drug (IC50 = 0.023 and 3.4 µM). Compounds 14 and 10 showed the best inhibitory activity toward AChE (IC50 = 0.71 µM) and BuChE (IC50 = 0.006 µM), respectively. The ligand-protein docking simulations and kinetic studies revealed that compound 14 and 10 could bind effectively to the peripheral anionic binding site (PAS) of the AChE and BuChE through mixed-type inhibition. In addition, the most potent compounds showed acceptable neuroprotective activity on H2O2- and Aβ-induced .neurotoxicity in PC12 cells, more than standard drugs. The compounds could block effectively self- and AChE-induced Aβ aggregation. All the results suggest that compounds 14 and 10 could be considered as promising multi-target-directed ligands against AD.
Collapse
|
28
|
Zhou X, Wang S, Zhang C, Lin Y, Lv J, Hu S, Zhang S, Li M. Colorimetric determination of amyloid-β peptide using MOF-derived nanozyme based on porous ZnO-Co 3O 4 nanocages. Mikrochim Acta 2021; 188:56. [PMID: 33502585 DOI: 10.1007/s00604-021-04705-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/09/2021] [Indexed: 12/24/2022]
Abstract
A sensitive and rapid colorimetric biosensor has been developed for determination of amyloid-β peptide (Aβ) and study of amyloidogenesis based on the high peroxidase-like activity of porous bimetallic ZnO-Co3O4 nanocages (NCs). Due to the high binding ability of Aβ monomer to ZnO-Co3O4 NCs, the catalytic activity of ZnO-Co3O4 NCs can be significantly suppressed by Aβ monomer. This finding forms the basis for a colorimetric assay for Aβ monomer detection. The detection limit for Aβ monomer is 3.5 nM with a linear range of 5 to 150 nM (R2 = 0.997). The system was successfully applied to the determination of Aβ monomer in rat cerebrospinal fluid. Critically, the different inhibition effects of monomeric and aggregated Aβ species on the catalytic activity of ZnO-Co3O4 NCs enabled the sensor to be used for tracking the dynamic progress of Aβ aggregation and screening Aβ inhibitors. Compared with the commonly used thioflavin T fluorescence assay, this method provided higher sensitivity to the formation of Aβ oligomer at the very early assembly stage. Our assay shows potential application in early diagnosis and therapy of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Xi Zhou
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuangling Wang
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Cong Zhang
- Department of Chemistry, School of Sciences, Hebei University of Science and Technology, Shijiazhuang, 050018, China.
| | - Yulong Lin
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jie Lv
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuyang Hu
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shanshan Zhang
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Meng Li
- College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
29
|
Xu S, Wang W, Dong X, Sun Y. Molecular Insight into Cu 2+-Induced Conformational Transitions of Amyloid β-Protein from Fast Kinetic Analysis and Molecular Dynamics Simulations. ACS Chem Neurosci 2021; 12:300-310. [PMID: 33401892 DOI: 10.1021/acschemneuro.0c00502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cu2+-mediated amyloid β-protein (Aβ) aggregation is implicated in the pathogenesis of Alzheimer's disease, so it is of significance to understand Cu2+-mediated conformational transitions of Aβ. Herein, four Aβ mutants were created by using the environment-sensitive cyanophenylalanine to respectively substitute F4, Y10, F19, and F20 residues of Aβ40. By using stopped-flow fluorescence spectroscopy and molecular dynamics (MD) simulations, the early stage conformational transitions of the mutants mediated by Cu2+ binding were investigated. The fast kinetics unveils that Cu2+ has more significant influence on the conformational changes of N-terminal (F4 and Y10) than on the central hydrophobic core (CHC, F19, and F20) under different pH conditions (pH 6.6-8.0), especially Y10. Interestingly, lag periods of the conformational transitions are observed for the F19 and F20 mutants at pH 8.0, indicating the slow response of the two mutation sites on the conformational transitions. More importantly, significantly longer lag periods for F20 than for F19 indicate the conduction of the transition from F19 to F20. The conduction time (difference in lag period) decreases from 4.5 s at Cu2+ = 0 to undetectable (<1 ms) at Cu2+ = 10 μM. The significant difference in the response time of F19 and F20 and the fast local conformational changes of Y10 imply that the conformational transitions of Aβ start around Y10. MD simulations support the observation of hydrophobicity increase at N-terminal during the conformational transitions of Aβ-Cu2+. It also reveals that Y10 is immediately approached by Cu2+, supporting the speculation that the starting point of conformational transitions of Aβ is near Y10. The work has provided molecular insight into the early stage conformational transitions of Aβ40 mediated by Cu2+.
Collapse
Affiliation(s)
- Shaoying Xu
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Wenjuan Wang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| |
Collapse
|
30
|
Wang H, Xu X, Pan YC, Yan Y, Hu XY, Chen R, Ravoo BJ, Guo DS, Zhang T. Recognition and Removal of Amyloid-β by a Heteromultivalent Macrocyclic Coassembly: A Potential Strategy for the Treatment of Alzheimer's Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2006483. [PMID: 33325586 DOI: 10.1002/adma.202006483] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/20/2020] [Indexed: 06/12/2023]
Abstract
The imbalance of amyloid-β (Aβ) production and clearance causes aggregation of Aβ1-42 monomers to form fibrils and amyloid plaques, which is an indispensable process in the pathogenesis of Alzheimer's disease (AD), and eventually leads to pathological changes and cognitive impairment. Consequently, Aβ1-42 is the most important target for the treatment of AD. However, developing a single treatment method that can recognize Aβ1-42 , inhibit Aβ1-42 fibrillation, eliminate amyloid plaques, improve cognitive impairments, and alleviate AD-like pathology is challenging. Here, a coassembly composed of cyclodextrin (CD) and calixarene (CA) is designed, and it is used as an anti-Aβ therapy agent. The CD-CA coassembly is based on the previously reported heteromultivalent recognition strategy and is able to successfully eliminate amyloid plaques and degrade Aβ1-42 monomers in 5xFAD mice. More importantly, the coassembly improves recognition and spatial cognition deficits, and synaptic plasticity impairment in the 5xFAD mice. In addition, the coassembly ameliorates AD-like pathology including prevention of neuronal apoptosis and oxidant stress, and alteration of M1/M2 microglial polarization states. This supramolecular approach makes full use of both molecular recognition and self-assembly of macrocyclic amphiphiles, and is a promising novel strategy for AD treatment.
Collapse
Affiliation(s)
- Hui Wang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - XinXin Xu
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Yu-Chen Pan
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Tianjin, 300071, P. R. China
| | - YuXing Yan
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Xin-Yue Hu
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Tianjin, 300071, P. R. China
| | - RunWen Chen
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Bart Jan Ravoo
- Organic Chemistry Institute and Center for Soft Nanoscience (SoN), Westfälische Wilhelms-Universität Münster, Busso-Peus-Straße 10, Münster, 48149, Germany
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Tianjin, 300071, P. R. China
| | - Tao Zhang
- College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
31
|
Antonoglou O, Giannousi K, Mourdikoudis S, Dendrinou-Samara C. Magnetic nanoemulsions as candidates for Alzheimer's disease dual imaging theranostics. NANOTECHNOLOGY 2020; 31:465702. [PMID: 32750688 DOI: 10.1088/1361-6528/abac35] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia linked to the accumulation of amyloid-beta (Aβ) plaques-fibrils that impair cognitive functions. Magnetic nanoparticles (MNPs) are emerging as promising tools for the crusade against AD owning to appropriate biocompatibility and facile functionalization that can lead to theranostic agents. Herein, the fabrication of a multimodal (magnetic resonance imaging (MRI), fluorescence imaging, and drug carrier) magnetic nanoemulsion (MNE) is reported as an AD theranostic candidate. Initially zinc ferrite MNPs of high saturation magnetization (129 emu g-1) were synthesized through a modified microwave-assisted polyol process. Memantine (a registered AD drug) was labeled with fluorescein (Mem-Flu) and encapsulated with the MNPs in sodium dodecyl sulfate micelles to form the MNE. Small hydrodynamic size (107), high encapsulation (77.5%) and loading efficiencies (86.1%) and sufficient transverse relaxivity (48.7 mM-1 s-1) were achieved through the design while sustained release of Mem-Flu was unveiled by in zero-order, first-order, Higuchi and Korsmeyer-Peppas pharmacokinetic models. Moreover, the MNE acquired fluorescence imaging ability of Aβ1-42 peptide monomers and/or plaques-fibrils via the fluorescein labeling of Memantine. A novel inorganic-organic hybrid multimodal AD theranostic candidate is presented.
Collapse
Affiliation(s)
- Orestis Antonoglou
- Laboratory of Inorganic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | | | | | | |
Collapse
|
32
|
Song W, Jing Z, Meng L, Zhou R. Tungsten Oxide Nanodots Exhibit Mild Interactions with WW and SH3 Modular Protein Domains. ACS OMEGA 2020; 5:11005-11012. [PMID: 32455221 PMCID: PMC7241039 DOI: 10.1021/acsomega.0c00822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/29/2020] [Indexed: 06/11/2023]
Abstract
Tungsten oxide nanodot (WO3-x ) is an active photothermal nanomaterial that has recently been discovered as a promising candidate for tumor theranostics and treatments. However, its potential cytotoxicity remains elusive and needs to be evaluated to assess its biosafety risks. Herein, we investigate the interactions between WO3-x and two ubiquitous protein domains involved in protein-protein interactions, namely, WW and SH3 domains, using atomistic molecular dynamics simulations. Our results show that WO3-x interacts only weakly with the key residues at the putative proline-rich motif (PRM) ligand-binding site of both domains. More importantly, our free energy landscape calculations reveal that the binding strength between WO3-x and WW/SH3 is weaker than that of the native PRM ligand with WW/SH3, implying that WO3-x has a limited inhibitory effect over PRM on both the WW and SH3 domains. These findings suggest that the cytotoxic effects of WO3-x on the key modular protein domains could be very mild, which provides new insights for the future potential biomedical applications of this nanomaterial.
Collapse
Affiliation(s)
- Wei Song
- Institute of Quantitative
Biology, Zhejiang University, Hangzhou 310027, China
| | - Zhifeng Jing
- IBM Thomas J. Watson Research Center, Yorktown Heights, New York 10598, United States
| | - Lijun Meng
- Institute of Quantitative
Biology, Zhejiang University, Hangzhou 310027, China
| | - Ruhong Zhou
- Institute of Quantitative
Biology, Zhejiang University, Hangzhou 310027, China
- IBM Thomas J. Watson Research Center, Yorktown Heights, New York 10598, United States
| |
Collapse
|
33
|
Waku T, Kobayashi Y, Wada M, Hamawaki T, Handa A, Okuda M, Sugimoto H, Kobori A, Tanaka N. Inhibition of Amyloid β Fibrillation by Nanoparticles Composed of Ovalbumin-derived Amphiphilic Peptides. CHEM LETT 2020. [DOI: 10.1246/cl.200048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Tomonori Waku
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Gosyokaido-cho, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Yukako Kobayashi
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Gosyokaido-cho, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Mei Wada
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Gosyokaido-cho, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Taiga Hamawaki
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Gosyokaido-cho, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Akihiro Handa
- R & D Division, Kewpie Corporation, 2-5-7 Sengawa-cho, Chofu, Tokyo 182-0002, Japan
| | - Michiaki Okuda
- Greentech Co. Ltd., 1-7-7 Yaesu, Chuo-ku, Tokyo 103-0028, Japan
| | - Hachiro Sugimoto
- Greentech Co. Ltd., 1-7-7 Yaesu, Chuo-ku, Tokyo 103-0028, Japan
- Faculty of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto 610-0394, Japan
| | - Akio Kobori
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Gosyokaido-cho, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Naoki Tanaka
- Faculty of Molecular Chemistry and Engineering, Kyoto Institute of Technology, Gosyokaido-cho, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| |
Collapse
|
34
|
Liu W, Wang W, Dong X, Sun Y. Near-Infrared Light-Powered Janus Nanomotor Significantly Facilitates Inhibition of Amyloid-β Fibrillogenesis. ACS APPLIED MATERIALS & INTERFACES 2020; 12:12618-12628. [PMID: 32105446 DOI: 10.1021/acsami.0c02342] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Inspired by the natural motors, artificial nanomotors (NMs) have emerged as intelligent, advanced, and multifunctional nanoplatforms that can perform complex tasks in living environments. However, the functionalization of these fantastic materials is in its infancy, hindering the success of this booming field. Herein, an inhibitor-conjugated near-infrared (NIR) laser-propelled Janus nanomotor (JNM-I) was constructed and first applied in the modulation of amyloid-β protein (Aβ) aggregation which is highly associated with Alzheimer's disease (AD). Under NIR light illumination, JNM-I exhibited efficient propulsion through the "self-thermophoresis" effect, and the active motion of JNM-I increased the opportunity of the contacts between the immobilized inhibitors and Aβ species, leading to an intensification of JNM-I on modulating the on-pathway Aβ aggregation, as evidenced by the distinct changes of the amyloid morphology, conformation, and cytotoxicity. For example, with a NIR irradiation, 200 μg/mL of JNM-I increased the cultured SH-SY5Y cell viability from 68% to nearly 100%, but it only protected the cells to 89% viability without an NIR irradiation. Meanwhile, the NIR irradiation effectively improved the blood-brain barrier (BBB) penetration of JNM-I. Such a JNM-I has connected artificial nanomotors with protein aggregation and provided new insight into the potential applications of various nanomotors in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Wei Liu
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Wenjuan Wang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| |
Collapse
|
35
|
Li G, Yang W, Li W, Luo Y, Lim Y, Li Y, Paul A, Segal D, Hong L, Li Y. Rational Design of a Cocktail of Inhibitors against Aβ Aggregation. Chemistry 2020; 26:3499-3503. [DOI: 10.1002/chem.201905621] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/21/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Gao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
- Institute of OceanographyMinjiang University 350108 Fuzhou P. R. China
| | - Wu‐Yue Yang
- Zhou Pei-Yuan Center for Applied MathematicsTsinghua University 100084 Beijing P. R. China
| | - Wen‐Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
| | - Yun‐Yi Luo
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
| | - Yeh‐Jun Lim
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
| | - Yang Li
- Institute for Science and TechnologyShandong University of, Traditional Chinese Medicine 250355 Jinan P. R. China
| | - Ashim Paul
- School of Molecular Microbiology & BiotechnologyTel Aviv University 69978 Tel Aviv Israel
| | - Daniel Segal
- School of Molecular Microbiology & BiotechnologyTel Aviv University 69978 Tel Aviv Israel
- Sagol Interdisciplinary School of NeurosciencesTel Aviv University 69978 Tel Aviv Israel
| | - Liu Hong
- Zhou Pei-Yuan Center for Applied MathematicsTsinghua University 100084 Beijing P. R. China
| | - Yan‐Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
- Beijing Institute for Brain Disorders 100069 Beijing P. R. China
- Center for Synthetic and Systems BiologyTsinghua University 100084 Beijing P. R. China
| |
Collapse
|
36
|
Gupta S, Dasmahapatra AK. Caffeine destabilizes preformed Aβ protofilaments: insights from all atom molecular dynamics simulations. Phys Chem Chem Phys 2019; 21:22067-22080. [PMID: 31565708 DOI: 10.1039/c9cp04162a] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aggregation and deposition of neurotoxic Aβ fibrils are key in the etiology of Alzheimer's disease (AD). It has been clinically recognized as a major form of dementia across the globe. Finding and testing various natural compounds to target Aβ fibrils to disrupt their stable structures seems to be a promising and attractive therapeutic strategy. The destabilization effects of caffeine on Aβ fibrils are investigated via in silico studies, where a series of molecular dynamics (MD) simulations, each of 100 ns, was conducted. The simulation outcomes obtained henceforth clearly indicated the drift of the terminal chains from the protofibrils, leading to disorganization of the characteristically organized cross-β structures of Aβ fibrils. The structural instability of Aβ17-42 protofibrils is explained through enhanced fluctuations in the RMSD, radius of gyration and RMSF values in the presence of caffeine. The key interactions providing stability, comprising D23-K28 salt bridges, intra- and inter-chain hydrogen bonding and hydrophobic interactions involving interchain A21-V36 and F19-G38 and intrachain L34-V36, were found to be disrupted due to increases in the distances between the participating components. The loss of β-sheet structure with the introduction of turns and α-helices in terminal chains may further inhibit the formation of higher order aggregates, which is necessary to stop the progression of the disease. The atomistic details obtained via MD studies relating to the mechanism behind the underlying destabilization of Aβ17-42 protofibrils by caffeine encourage further investigations exploring the potency of natural compounds to treat AD via disrupting preformed neurotoxic Aβ protofibrils.
Collapse
Affiliation(s)
- Shivani Gupta
- Department of Chemical Engineering and Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India.
| | | |
Collapse
|
37
|
Asadbegi M, Shamloo A. Identification of a Novel Multifunctional Ligand for Simultaneous Inhibition of Amyloid-Beta (Aβ 42) and Chelation of Zinc Metal Ion. ACS Chem Neurosci 2019; 10:4619-4632. [PMID: 31566950 DOI: 10.1021/acschemneuro.9b00468] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Zinc binding to β-amyloid structure could promote amyloid-β aggregation, as well as reactive oxygen species (ROS) production, as suggested in many experimental and theoretical studies. Therefore, the introduction of multifunctional drugs capable of chelating zinc metal ion and inhibiting Aβ aggregation is a promising strategy in the development of AD treatment. The present study has evaluated the efficacy of a new bifunctional peptide drug using molecular docking and molecular dynamics (MD) simulations. This drug comprises two different domains, an inhibitor domain, obtained from the C-terminal hydrophobic region of Aβ, and a Zn2+ chelating domain, derived from rapeseed meal, merge with a linker. The multifunctionality of the ligand was evaluated using a comprehensive set of MD simulations spanning up to 3.2 μs including Aβ relaxation, ligand-Zn2+ bilateral interaction, and, more importantly, ligand-Zn2+-Aβ42 trilateral interactions. Analysis of the results strongly indicated that the bifunctional ligand can chelate zinc metal ion and avoid Aβ aggregation simultaneously. The present study illustrated that the proposed ligand has considerable hydrophobic interactions and hydrogen bonding with monomeric Aβ in the presence of zinc metal ion. Therefore, in light of these considerable interactions and contacts, the α-helical structure of Aβ has been enhanced, while the β-sheet formation is prevented and the α-helix native structure is protected. Furthermore, the analysis of interactions between Aβ and ligand-zinc complex revealed that the zinc metal ion is coordinated to Met13, the ending residue of the ligand and merely one residue in Aβ. The results have proven the previous experimental and theoretical findings in the literature about Aβ interactions with zinc metal ion and also Aβ interactions with the first domain of the proposed ligand. Moreover, the current research has evaluated the chelation using MD simulation and linear interaction energy (LIE) methods, and the result has been satisfactorily verified with previous experimental and theoretical (DFT) studies.
Collapse
Affiliation(s)
- Mohsen Asadbegi
- Sharif University of Technology, School of Mechanical Engineering, Tehran 94305, Iran
| | - Amir Shamloo
- Sharif University of Technology, School of Mechanical Engineering, Tehran 94305, Iran
| |
Collapse
|
38
|
Jokar S, Khazaei S, Behnammanesh H, Shamloo A, Erfani M, Beiki D, Bavi O. Recent advances in the design and applications of amyloid-β peptide aggregation inhibitors for Alzheimer's disease therapy. Biophys Rev 2019; 11:10.1007/s12551-019-00606-2. [PMID: 31713720 DOI: 10.1007/s12551-019-00606-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/31/2019] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) is an irreversible neurological disorder that progresses gradually and can cause severe cognitive and behavioral impairments. This disease is currently considered a social and economic incurable issue due to its complicated and multifactorial characteristics. Despite decades of extensive research, we still lack definitive AD diagnostic and effective therapeutic tools. Consequently, one of the most challenging subjects in modern medicine is the need for the development of new strategies for the treatment of AD. A large body of evidence indicates that amyloid-β (Aβ) peptide fibrillation plays a key role in the onset and progression of AD. Recent studies have reported that amyloid hypothesis-based treatments can be developed as a new approach to overcome the limitations and challenges associated with conventional AD therapeutics. In this review, we will provide a comprehensive view of the challenges in AD therapy and pathophysiology. We also discuss currently known compounds that can inhibit amyloid-β (Aβ) aggregation and their potential role in advancing current AD treatments. We have specifically focused on Aβ aggregation inhibitors including metal chelators, nanostructures, organic molecules, peptides (or peptide mimics), and antibodies. To date, these molecules have been the subject of numerous in vitro and in vivo assays as well as molecular dynamics simulations to explore their mechanism of action and the fundamental structural groups involved in Aβ aggregation. Ultimately, the aim of these studies (and current review) is to achieve a rational design for effective therapeutic agents for AD treatment and diagnostics.
Collapse
Affiliation(s)
- Safura Jokar
- Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. BOX: 14155-6559, Tehran, Iran
| | - Saeedeh Khazaei
- Department of Pharmaceutical Biomaterials , Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. BOX: 14155-6559, Tehran, Iran
| | - Hossein Behnammanesh
- Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, P.O. BOX: 14155-6559, Tehran, Iran
| | - Amir Shamloo
- Department of Mechanical Engineering, Sharif University of Technology, P.O. Box: 11365-11155, Tehran, Iran
| | - Mostafa Erfani
- Radiation Application Research School, Nuclear Science and Technology Research Institute (NSTRI), P.O. Box: 14155-1339, Tehran, Iran
| | - Davood Beiki
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, P.O. BOX: 14155-6559, Tehran, Iran
| | - Omid Bavi
- Department of Mechanical and Aerospace Engineering, Shiraz University of Technology, P.O. Box: 71555-313, Shiraz, Iran.
| |
Collapse
|
39
|
Jiang X, Halmes AJ, Licari G, Smith JW, Song Y, Moore EG, Chen Q, Tajkhorshid E, Rienstra CM, Moore JS. Multivalent Polymer-Peptide Conjugates-A General Platform for Inhibiting Amyloid Beta Peptide Aggregation. ACS Macro Lett 2019; 8:1365-1371. [PMID: 32149017 PMCID: PMC7059649 DOI: 10.1021/acsmacrolett.9b00559] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Protein aggregation is implicated in multiple deposition diseases including Alzheimer's Disease, which features the formation of toxic aggregates of amyloid beta (Aβ) peptides. Many inhibitors have been developed to impede or reverse Aβ aggregation. Multivalent inhibitors, however, have been largely overlooked despite the promise of high inhibition efficiency endowed by the multivalent nature of Aβ aggregates. In this work, we report the success of multivalent polymer-peptide conjugates (mPPCs) as a general class of inhibitors of the aggregation of Aβ40. Significantly delayed onset of fibril formation was realized using mPPCs prepared from three peptide/peptoid ligands covering a range of polymer molecular weights (MWs) and ligand loadings. Dose dependence studies showed that the nature of the ligands is a key factor in mPPC inhibition potency. The negatively charged ligand LPFFD (LD) leads to more efficient mPPCs compared to the neutral ligands, and is most effective at 7% ligand loading across different MWs. Molecular dynamics simulations along with dynamic light scattering experiments suggest that mPPCs form globular structures in solution due to ligand-ligand interactions. Such interactions are key to the spatial proximity of ligands and thus to the multivalency effect of mPPC inhibition. Excess ligand-ligand interactions, however, reduce the accessibility of mPPC ligands to Aβ peptides, and impair the overall inhibition potency.
Collapse
Affiliation(s)
- Xing Jiang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
| | - Abigail J Halmes
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Giuseppe Licari
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
| | - John W Smith
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yang Song
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Edwin G Moore
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Qian Chen
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Emad Tajkhorshid
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Chad M Rienstra
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jeffrey S Moore
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
40
|
Narang SS, Shuaib S, Goyal D, Goyal B. In silico-guided identification of potential inhibitors against β2m aggregation in dialysis-related amyloidosis. J Biomol Struct Dyn 2019; 38:3927-3941. [DOI: 10.1080/07391102.2019.1668852] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Simranjeet Singh Narang
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, India
| | - Suniba Shuaib
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, India
| | - Deepti Goyal
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, Punjab, India
| | - Bhupesh Goyal
- School of Chemistry and Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab, India
| |
Collapse
|
41
|
Kaur A, Narang SS, Kaur A, Mann S, Priyadarshi N, Goyal B, Singhal NK, Goyal D. Multifunctional Mono-Triazole Derivatives Inhibit Aβ42 Aggregation and Cu2+-Mediated Aβ42 Aggregation and Protect Against Aβ42-Induced Cytotoxicity. Chem Res Toxicol 2019; 32:1824-1839. [DOI: 10.1021/acs.chemrestox.9b00168] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Amandeep Kaur
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib 140406, Punjab, India
| | - Simranjeet Singh Narang
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib 140406, Punjab, India
| | - Anupamjeet Kaur
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib 140406, Punjab, India
| | - Sukhmani Mann
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib 140406, Punjab, India
| | - Nitesh Priyadarshi
- National Agri-Food Biotechnology Institute, S.A.S. Nagar 140306, Punjab, India
| | - Bhupesh Goyal
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147004, Punjab, India
| | - Nitin Kumar Singhal
- National Agri-Food Biotechnology Institute, S.A.S. Nagar 140306, Punjab, India
| | - Deepti Goyal
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib 140406, Punjab, India
| |
Collapse
|
42
|
Wang W, Dong X, Sun Y. Modification of Serum Albumin by High Conversion of Carboxyl to Amino Groups Creates a Potent Inhibitor of Amyloid β-Protein Fibrillogenesis. Bioconjug Chem 2019; 30:1477-1488. [PMID: 30964649 DOI: 10.1021/acs.bioconjchem.9b00209] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Fibrillogenesis of amyloid β-protein (Aβ) has been thought to be implicated in the progression of Alzheimer's disease (AD). Therefore, development of high-efficiency inhibitors is one of the strategies for the prevention and treatment of AD. Serum albumin has been found to capture Aβ monomers through its hydrophobic groove and suppress amyloid formation, but the inhibition efficiency is limited. Inspired by the strong inhibition potency of a basic protein, human lysozyme, we have herein proposed to develop a basified serum albumin by converting carboxyl groups into amino groups with ethylenediamine conjugated on the protein surface. The idea was verified with both bovine and human serum albumins (BSA/HSA). Four basified BSA (BSA-B) preparations with amino modification degrees (MDs) from 8.0 to 41.5 were first synthesized. Extensive biophysical and biological analyses revealed that the inhibition potency significantly increased with increasing amino MD. BSA-B of the highest MD (41.5), BSA-B4, which had an isoelectric point of 9.7, presented strong inhibition on Aβ42 fibrillation at a concentration as low as 0.5 μM, at which it functioned similarly with 25 μM native BSA to impede 25 μM Aβ fibrillation. Cell viability assays also confirmed that the detoxification of 5 μM BSA-B4 was superior over 25 μM native BSA by increasing cell viability from 60.6% to 96.0%. Fluorescence quenching study unveiled the decrease of the binding affinity between Aβ42 and the hydrophobic pocket region of BSA-B4, while quartz crystal microbalance experiments demonstrated that the binding constant of BSA-B4 to Aβ42 increased nearly 5 times. Therefore, the increase of electrostatic interactions between BSA-B4 and Aβ42 was the main reason for its high potency. Hence, aminated BSA achieved a conversion of binding way to Aβ from a mainly single-site hydrophobic binding to multiregional electrostatic interactions. Similar results were obtained with basified HSA preparations on inhibiting the amyloid formation and cytotoxicity. This work has thus provided new insights into the development of more efficient protein-based inhibitors against Aβ fibrillogenesis.
Collapse
Affiliation(s)
- Wenjuan Wang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
| | - Xiaoyan Dong
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology , Tianjin University , Tianjin 300072 , China
| |
Collapse
|
43
|
Liu W, Dong X, Sun Y. d-Enantiomeric RTHLVFFARK-NH 2: A Potent Multifunctional Decapeptide Inhibiting Cu 2+-Mediated Amyloid β-Protein Aggregation and Remodeling Cu 2+-Mediated Amyloid β Aggregates. ACS Chem Neurosci 2019; 10:1390-1401. [PMID: 30650306 DOI: 10.1021/acschemneuro.8b00440] [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] [Indexed: 12/12/2022] Open
Abstract
Aggregation of amyloid β-protein (Aβ) into β-sheet-rich plaques is a general feature of Alzheimer's disease (AD). Homeostasis dysregulation of Cu2+ mediates Aβ to form high cytotoxic aggregates, which causes cell damage by generation of reactive oxygen species (ROS). To improve the inhibitory potency and explore the multifaceted functions of our previously designed decapeptide, RTHLVFFARK-NH2 (RK10), we have herein reformulated the decapeptide into its d-enantiomer, rk10, and the effects of chirality on Aβ aggregation, Cu2+-mediated Aβ aggregations, and aggregate-remodeling effects were investigated. The results revealed the following: (1) The d-enantiomer presented enhanced inhibitory potency on Aβ fibrillogenesis in comparison to RK10; rk10 and RK10 increased the cell viability from 60% to 91% and 71%, respectively, at an equimolar concentration to Aβ. (2) The enantiomers were chemically equivalent to Cu2+ chelation, ROS suppression and oxidative damage rescue. (3) The d-enantiomer exhibited higher performance to inhibit Cu2+-mediated Aβ aggregation, and more significantly attenuated the cytotoxicity caused by Aβ42-Cu2+ complex than RK10. Cell viability was rescued from 51% to 89% and 74% by coincubating with rk10 and RK10 at 50 μM, respectively. Intracellular ROS levels generated by Aβ42 and Aβ42-Cu2+ species were also remarkably decreased by treating with rk10. (4) The enantiomers could remodel mature Aβ42-Cu2+ aggregates by Cu2+ chelation, and rk10 showed higher performance than RK10, as evidenced by the enhanced cell viability from 57% to 86% by RK10 and to 96% by rk10. The d-enantiomer also showed higher ability than RK10 on protecting the disrupted species from reaggregation. Taken together, D-chiral derivatization of the decapeptide resulted in a potent multifunctional agent in inhibiting Cu2+-mediated Aβ aggregation and remodeling mature Aβ-Cu2+ species. To the best of our knowledge, this is the first investigation on the chirality effect of a multifunctional peptide inhibitor on Cu2+-mediated Aβ aggregation and on the remodeling effect of mature Aβ-Cu2+ aggregates. The work provides new insights into the critical role of chirality in the multifaceted functions of peptide inhibitors against amyloid formation and its toxicity.
Collapse
Affiliation(s)
- Wei Liu
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| |
Collapse
|
44
|
Lu J, Cao Q, Wang C, Zheng J, Luo F, Xie J, Li Y, Ma X, He L, Eisenberg D, Nowick J, Jiang L, Li D. Structure-Based Peptide Inhibitor Design of Amyloid-β Aggregation. Front Mol Neurosci 2019; 12:54. [PMID: 30886570 PMCID: PMC6409328 DOI: 10.3389/fnmol.2019.00054] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/12/2019] [Indexed: 12/20/2022] Open
Abstract
Many human neurodegenerative diseases are associated with amyloid fibril formation. Inhibition of amyloid formation is of importance for therapeutics of the related diseases. However, the development of selective potent amyloid inhibitors remains challenging. Here based on the structures of amyloid β (Aβ) fibrils and their amyloid-forming segments, we designed a series of peptide inhibitors using RosettaDesign. We further utilized a chemical scaffold to constrain the designed peptides into β-strand conformation, which significantly improves the potency of the inhibitors against Aβ aggregation and toxicity. Furthermore, we show that by targeting different Aβ segments, the designed peptide inhibitors can selectively recognize different species of Aβ. Our study developed an approach that combines the structure-based rational design with chemical modification for the development of amyloid inhibitors, which could be applied to the development of therapeutics for different amyloid-related diseases.
Collapse
Affiliation(s)
- Jinxia Lu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Cao
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Chuchu Wang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jing Zheng
- Shanghai Center for Women and Children's Health, Shanghai, China
| | - Feng Luo
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jingfei Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yichen Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojuan Ma
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lin He
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Center for Women and Children's Health, Shanghai, China
| | - David Eisenberg
- UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles, Los Angeles, CA, United States
| | - James Nowick
- Department of Chemistry, University of California, Irvine, Irvine, CA, United States
| | - Lin Jiang
- Department of Neurology, Easton Center for Alzheimer's Disease Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Dan Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
45
|
Yu Y, Yin T, Peng Q, Kong L, Li C, Tang D, Yin X. Simultaneous Monitoring of Amyloid-β (Aβ) Oligomers and Fibrils for Effectively Evaluating the Dynamic Process of Aβ Aggregation. ACS Sens 2019; 4:471-478. [PMID: 30693761 DOI: 10.1021/acssensors.8b01493] [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] [Indexed: 02/01/2023]
Abstract
Herein, we provide a proof of concept for a novel strategy that targets the assessment of the aggregation of amyloid-β (Aβ) by simultaneously determining its oligomers (Aβo) and fibrils (Aβf) in one analytical system. By fabricating and combining two immunosensors for Aβo and Aβf, respectively, we constructed a two-channel electrochemical system. The ratio of Aβf to Aβo was calculated and taken as a possible criterion for evaluating the extent of aggregation. Thereby, the presence of and transformation between oligomers and fibrils were accurately probed by incubating the Aβ monomer for different times and then calculating the ratios of Aβf to Aβo. The applicability of this method was further validated by tracking the dynamic progress of Aβ aggregation in the cerebrospinal fluid and tissues of Alzheimer's disease (AD) rats, which revealed that the ratio of Aβf to Aβo in rat brain gradually increased with the progression of AD, which was indicative of the severity of peptide aggregation during this process. Overall, this study represents the first example of a quantitative strategy for precisely evaluating the aggregation process that is related to pathological events in AD brain.
Collapse
Affiliation(s)
- Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Tianxiao Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Qiwen Peng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Lingna Kong
- Department of Chemistry, East China Normal University, 500 Dongchuan Road, Shanghai 200241, P. R. China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Daoquan Tang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, P. R. China
| |
Collapse
|
46
|
Morsy A, Trippier PC. Amyloid-Binding Alcohol Dehydrogenase (ABAD) Inhibitors for the Treatment of Alzheimer’s Disease. J Med Chem 2018; 62:4252-4264. [DOI: 10.1021/acs.jmedchem.8b01530] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
- Center for Chemical Biology, Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| |
Collapse
|
47
|
Houghtaling J, List J, Mayer M. Nanopore-Based, Rapid Characterization of Individual Amyloid Particles in Solution: Concepts, Challenges, and Prospects. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1802412. [PMID: 30225962 DOI: 10.1002/smll.201802412] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/15/2018] [Indexed: 06/08/2023]
Abstract
Aggregates of misfolded proteins are associated with several devastating neurodegenerative diseases. These so-called amyloids are therefore explored as biomarkers for the diagnosis of dementia and other disorders, as well as for monitoring disease progression and assessment of the efficacy of therapeutic interventions. Quantification and characterization of amyloids as biomarkers is particularly demanding because the same amyloid-forming protein can exist in different states of assembly, ranging from nanometer-sized monomers to micrometer-long fibrils that interchange dynamically both in vivo and in samples from body fluids ex vivo. Soluble oligomeric amyloid aggregates, in particular, are associated with neurotoxic effects, and their molecular organization, size, and shape appear to determine their toxicity. This concept article proposes that the emerging field of nanopore-based analytics on a single molecule and single aggregate level holds the potential to account for the heterogeneity of amyloid samples and to characterize these particles-rapidly, label-free, and in aqueous solution-with regard to their size, shape, and abundance. The article describes the concept of nanopore-based resistive pulse sensing, reviews previous work in amyloid analysis, and discusses limitations and challenges that will need to be overcome to realize the full potential of amyloid characterization on a single-particle level.
Collapse
Affiliation(s)
- Jared Houghtaling
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Jonathan List
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Michael Mayer
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| |
Collapse
|
48
|
Wei Y, Xie Z, Bi J, Zhu Z. Anti-inflammatory effects of bone marrow mesenchymal stem cells on mice with Alzheimer's disease. Exp Ther Med 2018; 16:5015-5020. [PMID: 30542456 PMCID: PMC6257159 DOI: 10.3892/etm.2018.6857] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 09/13/2018] [Indexed: 12/28/2022] Open
Abstract
Anti-inflammatory effects of bone marrow mesenchymal stem cells (BMSCs) on mice with Alzheimer's disease (AD) were investigated. Twenty amyloid precursor protein (APP)/presenilin-1 (PS1) double transgenic mice were randomly divided into two groups: the AD control group and the stem cell treatment group. The normal control group consisted of 10 non-transgenic mice. The stem cell treatment group was injected with BMSCs, and the two control groups were given the same volume of normal saline. The Morris water maze test was used to compare the memory function of mice, and the relative expression levels of β-site APP cleaving enzyme 1 (BACE1) and α-2-macroglobulin (A2M) genes were detected by fluorescence quantitative polymerase chain reaction (qPCR). Amyloid β (Aβ)1–42 content in brain tissues of mice and inflammatory cytokines, interleukin (IL)-1, IL-2, IL-10, tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) were detected using enzyme-linked immunosorbent assay (ELISA). Compared with that in the AD control group, the escape latency in the water maze in the stem cell treatment group was shortened, the time of crossing the ring for the first time was decreased, but the frequency of crossing the ring was increased (P<0.05). Aβ1–42 content in the AD control group was higher than that in the stem cell treatment group and the normal control group (P<0.05). The relative expression level of BACE1 gene in the stem cell treatment group was lower than that in the AD control group (P<0.05), but that of A2M gene was increased (P<0.05). At 14 days after treatment, the contents of IL-1, IL-2, TNF-α and IFN-γ in blood in the stem cell treatment group were lower than those in the AD control group (P<0.05). Human BMSCs can ameliorate the symptoms of AD by decreasing the levels of inflammatory cytokines and regulating the expression of Aβ-related genes.
Collapse
Affiliation(s)
- Yan Wei
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Zhaohong Xie
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Jianzhong Bi
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Zhengyu Zhu
- Department of Neurology Medicine, Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
49
|
Wu W, Liang X, Xie G, Chen L, Liu W, Luo G, Zhang P, Yu L, Zheng X, Ji H, Zhang C, Yi W. Synthesis and Evaluation of Novel Ligustrazine Derivatives as Multi-Targeted Inhibitors for the Treatment of Alzheimer's Disease. Molecules 2018; 23:molecules23102540. [PMID: 30301153 PMCID: PMC6222487 DOI: 10.3390/molecules23102540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 12/22/2022] Open
Abstract
A series of novel ligustrazine derivatives 8a–r were designed, synthesized, and evaluated as multi-targeted inhibitors for anti-Alzheimer’s disease (AD) drug discovery. The results showed that most of them exhibited a potent ability to inhibit both ChEs, with a high selectivity towards AChE. In particular, compounds 8q and 8r had the greatest inhibitory abilities for AChE, with IC50 values of 1.39 and 0.25 nM, respectively, and the highest selectivity towards AChE (for 8q, IC50 BuChE/IC50 AChE = 2.91 × 106; for 8r, IC50 BuChE/IC50 AChE = 1.32 × 107). Of note, 8q and 8r also presented potent inhibitory activities against Aβ aggregation, with IC50 values of 17.36 µM and 49.14 µM, respectively. Further cellular experiments demonstrated that the potent compounds 8q and 8r had no obvious cytotoxicity in either HepG2 cells or SH-SY5Y cells, even at a high concentration of 500 μM. Besides, a combined Lineweaver-Burk plot and molecular docking study revealed that these compounds might act as mixed-type inhibitors to exhibit such effects via selectively targeting both the catalytic active site (CAS) and the peripheral anionic site (PAS) of AChEs. Taken together, these results suggested that further development of these compounds should be of great interest.
Collapse
Affiliation(s)
- Wenhao Wu
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Xintong Liang
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Guoquan Xie
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Langdi Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Weixiong Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Guolin Luo
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Peiquan Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Lihong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Xuehua Zheng
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Hong Ji
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Chao Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| | - Wei Yi
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China.
| |
Collapse
|
50
|
Ma M, Gao N, Sun Y, Du X, Ren J, Qu X. Redox-Activated Near-Infrared-Responsive Polyoxometalates Used for Photothermal Treatment of Alzheimer's Disease. Adv Healthc Mater 2018; 7:e1800320. [PMID: 29920995 DOI: 10.1002/adhm.201800320] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 04/20/2018] [Indexed: 12/30/2022]
Abstract
Adjustable structure, excellent physiochemical properties, and good biocompatibility render polyoxometalates (POMs) as a suitable drug agent for the treatment of Alzheimer's disease (AD). However, previous works using POMs against AD just focus on the inhibition of amyloid-β (Aβ) monomer aggregation. In consideration that both Aβ fibrils and reactive oxygen species (ROS) are closely associated with clinical development of AD symptoms, it would be more effective if POMs can disaggregate Aβ fibrils and eliminate ROS as well. Herein, a redox-activated near-infrared (NIR) responsive POMs-based nanoplaform (rPOMs@MSNs@copolymer) is developed with high photothermal effect and antioxidant activity. The rPOMs@MSNs@copolymer can generate local hyperthermia to disaggregate Aβ fibrils under NIR laser irradiation because of POMs (rPOMs) with strong NIR absorption. Furthermore, Aβ-induced ROS can be scavenged by the antioxidant activity of rPOMs. To the authors' knowledge, there is no report of using rPOMs for NIR photothermal treatment of AD. This work may promote the development of multifunctional inorganic agents for biomedical applications.
Collapse
Affiliation(s)
- Mengmeng Ma
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
- University of Science and Technology of China; Hefei Anhui 230026 China
| | - Nan Gao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| | - Yuhuan Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
- University of Science and Technology of China; Hefei Anhui 230026 China
| | - Xiubo Du
- College of Life Sciences and Oceanography; Shenzhen Key Laboratory of Microbial Genetic Engineering; Shenzhen University; Shenzhen 518060 China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences; Changchun Jilin 130022 China
| |
Collapse
|