301
|
Tran PT, Hoang VH, Lee J, Hien TTT, Tung NT, Ngo ST. In vitroandin silicodetermination of glutaminyl cyclase inhibitors. RSC Adv 2019; 9:29619-29627. [PMID: 35531555 PMCID: PMC9071946 DOI: 10.1039/c9ra05763c] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of neurodegenerative disease currently. It is widely accepted that AD is characterized by the self-assembly of amyloid beta (Aβ) peptides. The human glutaminyl cyclase (hQC) enzyme is characterized by association with Aβ peptide generation. The development of hQC inhibitors could prevent the self-aggregation of Aβ peptides, resulting in impeding AD. Utilizing structural knowledge of the hQC substrates and known hQC inhibitors, new heterocyclic and peptidomimetic derivatives were synthesized and were able to inhibit the hQC enzyme. The inhibiting abilities of these compounds were evaluated using a fluorometric assay. The binding mechanism at the atomic level was estimated using molecular docking, free energy perturbation, and quantum chemical calculation methods. The predicted log(BBB) and human intestinal absorption values indicated that these compounds are able to permeate the blood–brain barrier and be well-absorbed through the gastrointestinal tract. Overall, 5,6-dimethoxy-N-(3-(5-methyl-1H-imidazol-1-yl)propyl)-1H-benzo[d]imidazol-2-amine (1_2) was indicated as a potential drug for AD treatment. Rational design of new hQC inhibitors.![]()
Collapse
Affiliation(s)
- Phuong-Thao Tran
- Department of Pharmaceutical Chemistry
- Hanoi University of Pharmacy
- Hanoi
- Vietnam
| | - Van-Hai Hoang
- Institute of Research and Development
- Duy Tan University
- Da Nang 550000
- Vietnam
| | - Jeewoo Lee
- Laboratory of Medicinal Chemistry
- College of Pharmacy
- Seoul National University
- Seoul
- Korea
| | | | - Nguyen Thanh Tung
- Institute of Materials Science
- Vietnam Academy of Science and Technology
- Hanoi
- Vietnam
| | - Son Tung Ngo
- Laboratory of Theoretical and Computational Biophysics
- Ton Duc Thang University
- Ho Chi Minh City
- Vietnam
- Faculty of Applied Sciences
| |
Collapse
|
302
|
Qiao Q, Wei G, Yao D, Song Z. Formation of α-helical and β-sheet structures in membrane-bound human IAPP monomer and the resulting membrane deformation. Phys Chem Chem Phys 2019; 21:20239-20251. [DOI: 10.1039/c9cp03151k] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Upon adsorption on membrane, human IAPP monomer takes conformational changes from coils to α-helices and β-sheets. The helices inserted and β on surface cause different types of membrane deformation, implying two distinct aggregation mechanisms.
Collapse
Affiliation(s)
- Qin Qiao
- Digital Medical Research Center
- School of Basic Medical Sciences
- Fudan University
- Shanghai 200032
- China
| | - Guanghong Wei
- Department of Physics
- State Key Laboratory of Surface Physics
- Key Laboratory for Computational Physical Science (Ministry of Education)
- Fudan University
- Shanghai 200438
| | - Demin Yao
- Digital Medical Research Center
- School of Basic Medical Sciences
- Fudan University
- Shanghai 200032
- China
| | - Zhijian Song
- Digital Medical Research Center
- School of Basic Medical Sciences
- Fudan University
- Shanghai 200032
- China
| |
Collapse
|
303
|
Nie RZ, Dang MZ, Li KK, Peng JM, Du J, Zhang MY, Li CM. A-type EGCG dimer, a new proanthocyanidins dimer from persimmon fruits, interacts with the amino acid residues of Aβ40 which possessed high aggregation-propensity and strongly inhibits its amyloid fibrils formation. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
304
|
Rana M, Sharma AK. Cu and Zn interactions with Aβ peptides: consequence of coordination on aggregation and formation of neurotoxic soluble Aβ oligomers. Metallomics 2019; 11:64-84. [DOI: 10.1039/c8mt00203g] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The coordination chemistry of transition metal ions (Fe, Cu, Zn) with the amyloid-β (Aβ) peptides has attracted a lot of attention in recent years due to its repercussions in Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Monika Rana
- Department of Chemistry
- Central University of Rajasthan
- Ajmer 305817
- India
| | - Anuj Kumar Sharma
- Department of Chemistry
- Central University of Rajasthan
- Ajmer 305817
- India
| |
Collapse
|
305
|
Saini RK, Shuaib S, Goyal D, Goyal B. Insights into the inhibitory mechanism of a resveratrol and clioquinol hybrid against Aβ42 aggregation and protofibril destabilization: A molecular dynamics simulation study. J Biomol Struct Dyn 2018; 37:3183-3197. [DOI: 10.1080/07391102.2018.1511475] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Rajneet Kaur Saini
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, India
| | - Suniba Shuaib
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, India
| | - Deepti Goyal
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, India
| | - Bhupesh Goyal
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, India
| |
Collapse
|
306
|
Li X, Xie B, Sun Y. Basified Human Lysozyme: A Potent Inhibitor against Amyloid β-Protein Fibrillogenesis. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:15569-15577. [PMID: 30407837 DOI: 10.1021/acs.langmuir.8b03278] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The aggregation of amyloid β-proteins (Aβ) has been recognized as a key process in the pathogenesis of Alzheimer's disease (AD), so inhibiting Aβ aggregation is an important strategy to prevent the onset and treatment of AD. Our recent work indicated that decreasing the positive charges (or introducing negative charges) on human lysozyme (hLys) was unfavorable in keeping the inhibiting capability of hLys on Aβ aggregation. Therefore, we have herein proposed to basify hLys by conversion of the carboxyl groups into amino groups by modification with ethylene diamine. Basified hLys (Lys-B) preparations of three modification degrees (MDs), denoted as hLys-B1 (MD, 1.5), hLys-B2 (MD, 3.3), and hLys-B3 (MD, 4.4), were synthesized for modulating Aβ fibrillogenesis. The hLys-B preparations kept the stability and biocompatibility as native hLys did, whereas the inhibitory potency of hLys-B on Aβ fibrillogenesis increased with increasing MD. Cytotoxicity analysis showed that cell viability with 2.5 μM hLys-B3 increased from 62.5% (with 25 μM Aβ only) to 76.1%, similar to the case with 12.5 μM hLys (75.5%); cell viability with 6.25 μM hLys-B3 increased to 82.0%, similar to the case with 25 μM hLys (80.9%). The results indicate about four- to fivefold increase in the inhibition efficiency of hLys by the amino modification. Mechanistic analysis suggests that such a superior inhibitory capability of hLys-B was attributed to its more widely distributed positive charges, which promoted broad electrostatic interactions between Aβ and hLys-B. Thus, hLys-B suppressed the conformational transition of Aβ to β-sheet structures at low concentrations (e.g., 2.5 μM hLys-B3), leading to changes in the aggregation pathway and the formation of Aβ species with less cytotoxicity. The findings provided new insights into the development of more potent protein-based inhibitors against Aβ fibrillogenesis.
Collapse
Affiliation(s)
- Xi Li
- 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
| | - Baolong Xie
- 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
- Institute of Tianjin Seawater Desalination and Multipurpose Utilization, State Oceanic Administration (SOA) , Tianjin 300192 , 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
|
307
|
Guo C, Zhou HX. Fatty Acids Compete with Aβ in Binding to Serum Albumin by Quenching Its Conformational Flexibility. Biophys J 2018; 116:248-257. [PMID: 30580919 DOI: 10.1016/j.bpj.2018.11.3133] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/16/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023] Open
Abstract
Human serum albumin (HSA) has been identified as an important regulator of amyloid-β (Aβ) fibrillization both in blood plasma and in cerebrospinal fluid. Fatty acids bind to HSA, and high serum levels of fatty acids increase the risk of Alzheimer's disease. In vitro, fatty-acid-loaded HSA (FA·HSA) loses the protective effect against Aβ fibrillization, but the mechanism underlying the interference of fatty acids on Aβ-HSA interactions has been unclear. Here, we used molecular dynamics simulations to gain atomic-level insight on the weak binding of monomeric Aβ40 and Aβ42 peptides with apo and FA·HSA. Consistent with recent NMR data, C-terminal residues of the Aβ peptides have the highest propensities for interacting with apo HSA. Interestingly, the Aβ binding residues of apo and FA·HSA exhibit distinct patterns, which qualitatively correlate with backbone flexibility. In FA·HSA, both flexibilities and Aβ binding propensities are relatively even among the three domains. In contrast, in apo HSA, domain III shows the highest flexibility and is the primary target for Aβ binding. Specifically, deformation of apo HSA creates strong binding sites within subdomain IIIb, around the interface between subdomains IIIa and IIIb, and at the cleft between domains III and I. Therefore, much like disordered proteins, HSA can take advantage of flexibility in forming promiscuous interactions with partners, until the flexibility is quenched by fatty-acid binding. Our work explains the effect of fatty acids on Aβ-HSA binding and contributes to the understanding of HSA regulation of Aβ aggregation.
Collapse
Affiliation(s)
- Cong Guo
- Department of Physics and International Centre for Quantum and Molecular Structures, Shanghai University, Shanghai, China.
| | - Huan-Xiang Zhou
- Department of Chemistry and Department of Physics, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
308
|
The exploration of novel Alzheimer's therapeutic agents from the pool of FDA approved medicines using drug repositioning, enzyme inhibition and kinetic mechanism approaches. Biomed Pharmacother 2018; 109:2513-2526. [PMID: 30551512 DOI: 10.1016/j.biopha.2018.11.115] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 11/19/2018] [Accepted: 11/25/2018] [Indexed: 12/11/2022] Open
Abstract
Novel drug development is onerous, time consuming and overpriced process with particularly low success and relatively high enfeebling rates. To overcome this burden, drug repositioning approach is being used to predict the possible therapeutic effects of FDA approved drugs in different diseases. Herein, we designed a computational and enzyme inhibitory mechanistic approach to fetch the promising drugs from the pool of FDA approved drugs against AD. The binding interaction patterns and conformations of screened drugs within active region of AChE were confirmed through molecular docking profiles. The possible associations of selected drugs with AD genes were predicted by pharmacogenomics analysis and confirmed through data mining. The stability behaviour of docked complexes (Drugs-AChE) were checked by MD simulations. The possible therapeutic potential of repositioned drugs against AChE were checked by in vitro analysis. Taken together, Cinitapride displayed a comparable results with standard and can be used as possible therapeutic agent in the treatment of AD.
Collapse
|
309
|
Gera J, Szögi T, Bozsó Z, Fülöp L, Barrera EE, Rodriguez AM, Méndez L, Delpiccolo CM, Mata EG, Cioffi F, Broersen K, Paragi G, Enriz RD. Searching for improved mimetic peptides inhibitors preventing conformational transition of amyloid-β42 monomer. Bioorg Chem 2018; 81:211-221. [DOI: 10.1016/j.bioorg.2018.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 01/14/2023]
|
310
|
Tran L, Kaffy J, Ongeri S, Ha-Duong T. Binding Modes of a Glycopeptidomimetic Molecule on Aβ Protofibrils: Implication for Its Inhibition Mechanism. ACS Chem Neurosci 2018; 9:2859-2869. [PMID: 30025208 DOI: 10.1021/acschemneuro.8b00341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
We recently reported that a glycopeptidomimetic molecule significantly delays the fibrillization process of Aβ42 peptide involved in Alzheimer's disease. However, the binding mode of this compound, named 3β, was not determined at the atomic scale, hindering our understanding of its mechanism of action and impeding structure-based design of new inhibitors. In the present study, we performed molecular docking calculations and molecular dynamics simulations to investigate the most probable structures of 3β complexed with Aβ protofibrils. Our results show that 3β preferentially binds to an area of the protofibril surface that coincides with the protofibril dimerization interface observed in the solid-state NMR structure 5KK3 from the PDB. Based on these observations, we propose a model of the inhibition mechanism of Aβ fibrillization by compound 3β.
Collapse
Affiliation(s)
- Linh Tran
- BioCIS, Université Paris-Sud, CNRS, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Julia Kaffy
- BioCIS, Université Paris-Sud, CNRS, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Sandrine Ongeri
- BioCIS, Université Paris-Sud, CNRS, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| | - Tâp Ha-Duong
- BioCIS, Université Paris-Sud, CNRS, Université Paris-Saclay, 92290 Châtenay-Malabry, France
| |
Collapse
|
311
|
Thai NQ, Bednarikova Z, Gancar M, Linh HQ, Hu CK, Li MS, Gazova Z. Compound CID 9998128 Is a Potential Multitarget Drug for Alzheimer's Disease. ACS Chem Neurosci 2018; 9:2588-2598. [PMID: 29775277 DOI: 10.1021/acschemneuro.8b00091] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We have probed small molecule compound CID 9998128 as a potential multitarget drug for the Alzheimer's disease (AD) using in silico and in vitro experiments. By all-atom simulation and molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) method, we have demonstrated that this compound strongly binds to both amyloid β42 (Aβ42) fibrils and β-secretase, and the van der Waals interaction dominates over the electrostatic interaction in binding affinity. A detailed analysis at the atomic level revealed that indazole in CID 99998128 structure made a major contribution to instability of all studied complexes. In vitro experiments have shown that CID 9998128 inhibits the Aβ42 amyloid fibrillization and is capable to clear Aβ42 fibrils. Moreover, the compound dose-dependently decreases β-site amyloid precursor protein cleaving enzyme (BACE-1) activity with EC50 value in micromolar range. Thus, our study has revealed that CID 9998128 is a good candidate for AD treatment through preventing production of Aβ peptides and degrading their aggregates. For drug design, we predict that the chemical structure of potent AD multitarget inhibitors should not contain indazole.
Collapse
Affiliation(s)
- Nguyen Quoc Thai
- Institute for Computational Sciences and Technology, SBI building, Quang Trung Software City, Tan Chanh Hiep Ward, District 12, Ho Chi Minh City, Vietnam
- Dong Thap University, 783 Pham Huu Lau Street, Ward 6, Cao Lanh City, Dong Thap, Vietnam
- Biomedical Engineering Department, University of Technology -VNU HCM, 268 Ly Thuong Kiet Str., Distr. 10, Ho Chi Minh City, Vietnam
| | - Zuzana Bednarikova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, Kosice 040 01, Slovakia
| | - Miroslav Gancar
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, Kosice 040 01, Slovakia
| | - Huynh Quang Linh
- Biomedical Engineering Department, University of Technology -VNU HCM, 268 Ly Thuong Kiet Str., Distr. 10, Ho Chi Minh City, Vietnam
| | - Chin-Kun Hu
- Institute of Physics, Academia Sinica, Nankang, Taipei 11529, Taiwan
- Physics Division, National Center for Theretical Sciences, Hsinchu 30013, Taiwan
- Department of Physics, National Dong Hwa University, Hualien 97401, Taiwan
- Department of Systems Science, University of Schanghai for Science and Technology, Shanghai 200093, China
| | - Mai Suan Li
- Institute of Physics, Polish Academy of Sciences, Al. Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Zuzana Gazova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, Kosice 040 01, Slovakia
| |
Collapse
|
312
|
Carballo-Pacheco M, Ismail AE, Strodel B. On the Applicability of Force Fields To Study the Aggregation of Amyloidogenic Peptides Using Molecular Dynamics Simulations. J Chem Theory Comput 2018; 14:6063-6075. [PMID: 30336669 DOI: 10.1021/acs.jctc.8b00579] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Molecular dynamics simulations play an essential role in understanding biomolecular processes such as protein aggregation at temporal and spatial resolutions which are not attainable by experimental methods. For a correct modeling of protein aggregation, force fields must accurately represent molecular interactions. Here, we study the effect of five different force fields on the oligomer formation of Alzheimer's Aβ16-22 peptide and two of its mutants: Aβ16-22(F19V,F20V), which does not form fibrils, and Aβ16-22(F19L) which forms fibrils faster than the wild type. We observe that while oligomer formation kinetics depends strongly on the force field, structural properties, such as the most relevant protein-protein contacts, are similar between them. The oligomer formation kinetics obtained with different force fields differ more from each other than the kinetics between aggregating and nonaggregating peptides simulated with a single force field. We discuss the difficulties in comparing atomistic simulations of amyloid oligomer formation with experimental observables.
Collapse
Affiliation(s)
- Martín Carballo-Pacheco
- Institute of Complex Systems: Structural Biochemistry (ICS-6) , Forschungszentrum Jülich GmbH , 52425 Jülich , Germany.,AICES Graduate School , RWTH Aachen University , Schinkelstraße 2 , 52062 Aachen , Germany
| | - Ahmed E Ismail
- AICES Graduate School , RWTH Aachen University , Schinkelstraße 2 , 52062 Aachen , Germany.,Aachener Verfahrenstechnik, Faculty of Mechanical Engineering , RWTH Aachen University , Schinkelstraße 2 , 52062 Aachen , Germany
| | - Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry (ICS-6) , Forschungszentrum Jülich GmbH , 52425 Jülich , Germany.,Institute of Theoretical and Computational Chemistry , Heinrich Heine University Düsseldorf , Universitätstrasse 1 , 40225 Düsseldorf , Germany
| |
Collapse
|
313
|
Li H, Wang X, Yu H, Zhu J, Jin H, Wang A, Yang Z. Combining in vitro and in silico Approaches to Find New Candidate Drugs Targeting the Pathological Proteins Related to the Alzheimer's Disease. Curr Neuropharmacol 2018; 16:758-768. [PMID: 29086699 PMCID: PMC6080099 DOI: 10.2174/1570159x15666171030142108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 09/24/2017] [Accepted: 10/10/2017] [Indexed: 01/10/2023] Open
Abstract
Background: Alzheimer’s disease (AD) as the most common cause of dementia among older people has aroused the universal concern of the whole world. However, until now there is still none effective treatments. Consequently, the development of new drugs targeting this complicated brain disorder is urgent and needs more efforts. In this review, we detailed the current state of knowledge about new candidate drugs targeting the pathological proteins especially the drugs which are employed using the combined methods of in vitro and in silico. Methods: We looked up and reviewed online papers related to the pathogenesis and new drugs development of AD. Then, articles up to the requirements were respectively analyzed and summaried to provide the latest knowledge about the pathogenic effect and the new candidate drugs targeting Aβ and Tau proteins. Results: New candidate drugs targeting the Aβ include decreasing the production, promoting the clearence and preventing aggregation. However these drugs have mostly failed in Phase III clinical trial stage due to the unsuccessful of reversing cognition symptoms. As to tau protein, the prevention of tau aggregation and propagation is a promising strategy to synthesize/design mechanism-based drugs against tauopathies. Some candidate drugs are under research. Moreover, because of the complex pathogenesis of AD, multi-target drugs have also shed light on the treatment of AD. Conclusion: Given to the consecutive failure of Aβ-directed drugs and the feasibilities of tau-targeted therapy, more and more researchers suggested that the AD treatment should be moved from Aβ to tau or focused on considering the soluble form of Aβ and tau as a whole. Moreover, the novel in silico methods also have great potential in drug discovery, drug repositioning, virtual screening of chemical libraries. No matter how many difficulties and challenges in prevention and treatment of AD, we firmly believe that the effective and safe drugs will be found using the combined methods in the immediate future with the global effort.
Collapse
Affiliation(s)
- Hui Li
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaobing Wang
- Tumor Marker Research Center, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hongmei Yu
- China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jing Zhu
- College of Pharmacy, The Ohio State University, Columbus, Ohio, 43210, United States
| | - Hongtao Jin
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Aiping Wang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhaogang Yang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, 43210, United States
| |
Collapse
|
314
|
Tian B, Cheng C, Yue T, Lin N, Ren H. Chemical identification of the amyloid peptide aggregation-prone Al(III)-peptide complexes by resonance Raman signatures: A computational study. Chem Phys 2018. [DOI: 10.1016/j.chemphys.2018.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
315
|
Press-Sandler O, Miller Y. Molecular mechanisms of membrane-associated amyloid aggregation: Computational perspective and challenges. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1889-1905. [DOI: 10.1016/j.bbamem.2018.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 01/02/2023]
|
316
|
Lu L, Deng Y, Li X, Li H, Karniadakis GE. Understanding the Twisted Structure of Amyloid Fibrils via Molecular Simulations. J Phys Chem B 2018; 122:11302-11310. [PMID: 30106299 DOI: 10.1021/acs.jpcb.8b07255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Accumulation and aggregation of amyloid are associated with the pathogenesis of many human diseases, such as Alzheimer's disease and Type 2 diabetes mellitus. Therefore, a quantitative understanding of the molecular mechanisms causing different aggregated structures and biomechanical properties of amyloid fibrils could shed some light into the progression of these diseases. In this work, we develop coarse-grained molecular dynamics (CGMD) models to simulate the dynamic self-assembly of two types of amyloids (amylin and amyloid β (Aβ)). We investigate the structural and mechanical properties of different types of aggregated amyloid fibrils. Our simulations demonstrate that amyloid fibrils could result from longitudinal growth of protofilament bundles, confirming one of the hypotheses on the fibril formation. In addition, we find that the persistence length of amylin fibrils increases concurrently with their pitch length, suggesting that the bending stiffness of amylin fibrils becomes larger when the amylin fibrils are less twisted. Similar results are observed for Aβ fibrils. These findings quantify the connection between the structural and the biomechanical properties of the fibrils. The CGMD models developed in this work can be potentially used to examine efficacy of anti-aggregation drugs, which could help in developing new treatments.
Collapse
Affiliation(s)
- Lu Lu
- Division of Applied Mathematics , Brown University , Providence , Rhode Island 02912 , United States
| | - Yixiang Deng
- School of Engineering , Brown University , Providence , Rhode Island 02912 , United States
| | - Xuejin Li
- Division of Applied Mathematics , Brown University , Providence , Rhode Island 02912 , United States
| | - He Li
- Division of Applied Mathematics , Brown University , Providence , Rhode Island 02912 , United States
| | - George Em Karniadakis
- Division of Applied Mathematics , Brown University , Providence , Rhode Island 02912 , United States
| |
Collapse
|
317
|
Törnquist M, Michaels TCT, Sanagavarapu K, Yang X, Meisl G, Cohen SIA, Knowles TPJ, Linse S. Secondary nucleation in amyloid formation. Chem Commun (Camb) 2018; 54:8667-8684. [PMID: 29978862 DOI: 10.1039/c8cc02204f] [Citation(s) in RCA: 310] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nucleation of new peptide and protein aggregates on the surfaces of amyloid fibrils of the same peptide or protein has emerged in the past two decades as a major pathway for both the generation of molecular species responsible for cellular toxicity and for the autocatalytic proliferation of peptide and protein aggregates. A key question in current research is the molecular mechanism and driving forces governing such processes, known as secondary nucleation. In this context, the analogies with other self-assembling systems for which monomer-dependent secondary nucleation has been studied for more than a century provide a valuable source of inspiration. Here, we present a short overview of this background and then review recent results regarding secondary nucleation of amyloid-forming peptides and proteins, focusing in particular on the amyloid β peptide (Aβ) from Alzheimer's disease, with some examples regarding α-synuclein from Parkinson's disease. Monomer-dependent secondary nucleation of Aβ was discovered using a combination of kinetic experiments, global analysis, seeding experiments and selective isotope-enrichment, which pinpoint the monomer as the origin of new aggregates in a fibril-catalyzed reaction. Insights into driving forces are gained from variations of solution conditions, temperature and peptide sequence. Selective inhibition of secondary nucleation is explored as an effective means to limit oligomer production and toxicity. We also review experiments aimed at finding interaction partners of oligomers generated by secondary nucleation in an ongoing aggregation process. At the end of this feature article we bring forward outstanding questions and testable mechanistic hypotheses regarding monomer-dependent secondary nucleation in amyloid formation.
Collapse
Affiliation(s)
- Mattias Törnquist
- Lund University, Department of Biochemistry and Structural Biology, Chemical Centre, PO Box 124, SE221 00 Lund, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
318
|
Orteca G, Tavanti F, Bednarikova Z, Gazova Z, Rigillo G, Imbriano C, Basile V, Asti M, Rigamonti L, Saladini M, Ferrari E, Menziani MC. Curcumin derivatives and Aβ-fibrillar aggregates: An interactions' study for diagnostic/therapeutic purposes in neurodegenerative diseases. Bioorg Med Chem 2018; 26:4288-4300. [PMID: 30031653 DOI: 10.1016/j.bmc.2018.07.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/10/2018] [Accepted: 07/14/2018] [Indexed: 01/01/2023]
Abstract
Several neurodegenerative diseases, like Alzheimer's (AD), are characterized by amyloid fibrillar deposition of misfolded proteins, and this feature can be exploited for both diagnosis and therapy design. In this paper, structural modifications of curcumin scaffold were examined in order to improve its bioavailability and stability in physiological conditions, as well as its ability to interfere with β-amyloid fibrils and aggregates. The acid-base behaviour of curcumin derivatives, their pharmacokinetic stability in physiological conditions, and in vitro ability to interfere with Aβ fibrils at different incubation time were investigated. The mechanisms governing these phenomena have been studied at atomic level by means of molecular docking and dynamic simulations. Finally, biological activity of selected curcuminoids has been investigated in vitro to evaluate their safety and efficiency in oxidative stress protection on hippocampal HT-22 mouse cells. Two aromatic rings, π-conjugated structure and H-donor/acceptor substituents on the aromatic rings showed to be the sine qua non structural features to provide interaction and disaggregation activity even at very low incubation time (2h). Computational simulations proved that upon binding the ligands modify the conformational dynamics and/or interact with the amyloidogenic region of the protofibril facilitating disaggregation. Significantly, in vitro results on hippocampal cells pointed out protection against glutamate toxicity and safety when administered at low concentrations (1 μM). On the overall, in view of its higher stability in physiological conditions with respect to curcumin, of his rapid binding to fibrillar aggregates and strong depolymerizing activity, phtalimmide derivative K2F21 appeared a good candidate for both AD diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Giulia Orteca
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Francesco Tavanti
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Zuzana Bednarikova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 040 01 Kosice, Slovakia
| | - Zuzana Gazova
- Department of Biophysics, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, 040 01 Kosice, Slovakia
| | - Giovanna Rigillo
- Department of Life Sciences, University of Modena and Reggio Emilia, via G. Campi 213/D, 41125 Modena, Italy
| | - Carol Imbriano
- Department of Life Sciences, University of Modena and Reggio Emilia, via G. Campi 213/D, 41125 Modena, Italy
| | - Valentina Basile
- Department of Life Sciences, University of Modena and Reggio Emilia, via G. Campi 213/D, 41125 Modena, Italy
| | - Mattia Asti
- Nuclear Medicine Unit, Advanced Technology Department, AUSL - IRCCS Reggio Emilia, viale Amendola 2, 42122 Reggio Emilia, Italy
| | - Luca Rigamonti
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Monica Saladini
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| | - Erika Ferrari
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via G. Campi 103, 41125 Modena, Italy.
| | - Maria Cristina Menziani
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, via G. Campi 103, 41125 Modena, Italy
| |
Collapse
|
319
|
Ilie IM, Caflisch A. Disorder at the Tips of a Disease-Relevant Aβ42 Amyloid Fibril: A Molecular Dynamics Study. J Phys Chem B 2018; 122:11072-11082. [PMID: 29965774 DOI: 10.1021/acs.jpcb.8b05236] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We present a simulation study of the early events of peptide dissociation from a fibril of the Alzheimer's Aβ42 peptide. The fibril consists of layers of two adjacent Aβ42 peptides each folded in an S-shaped structure which has been determined by solid state NMR spectroscopy of a monomorphic disease-relevant species. Multiple molecular dynamics runs (16 at 310 K and 15 at 370 K) were carried out starting from an 18-peptide protofibril for a cumulative sampling of about 15 μs. The simulations show structural stability of the fibrillar core and an overall increase in the twist to about 3 degrees. The N-terminal segment 1-14 is disordered in all peptides. At both ends of the fibril, the central segment 21-29, which includes part of the β2 strand, dissociates in some of the simulations. The β1 and β3 strands, residues 15-20 and 35-41, respectively, are structurally stable. The transient binding of the N-terminal stretch to the β3 strand of the adjacent peptide at the tip is likely to contribute to the arrest phase of the stop-and-go mechanism.
Collapse
Affiliation(s)
- Ioana M Ilie
- Department of Biochemistry , University of Zürich , 8057 Zürich , Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry , University of Zürich , 8057 Zürich , Switzerland
| |
Collapse
|
320
|
Zhang M, Zheng J, Nussinov R, Ma B. Molecular Recognition between Aβ-Specific Single-Domain Antibody and Aβ Misfolded Aggregates. Antibodies (Basel) 2018; 7:E25. [PMID: 31544877 PMCID: PMC6640678 DOI: 10.3390/antib7030025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/12/2022] Open
Abstract
Aβ is the toxic amyloid polypeptide responsible for Alzheimer's disease (AD). Prevention and elimination of the Aβ misfolded aggregates are the promising therapeutic strategies for the AD treatments. Gammabody, the Aβ-Specific Single-domain (VH) antibody, recognizes Aβ aggregates with high affinity and specificity and reduces their toxicities. Employing the molecular dynamics simulations, we studied diverse gammabody-Aβ recognition complexes to get insights into their structural and dynamic properties and gammabody-Aβ recognitions. Among many heterogeneous binding modes, we focused on two gammabody-Aβ recognition scenarios: recognition through Aβ β-sheet backbone and on sidechain surface. We found that the gammabody primarily uses the complementarity-determining region 3 (CDR3) loop with the grafted Aβ sequence to interact with the Aβ fibril, while CDR1/CDR2 loops have very little contact. The gammabody-Aβ complexes with backbone binding mode are more stable, explaining the gammabody's specificity towards the C-terminal Aβ sequence.
Collapse
Affiliation(s)
- Mingzhen Zhang
- Department of Chemical & Biomolecular Engineering, the University of Akron, Akron, OH 44325, USA.
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA.
| | - Jie Zheng
- Department of Chemical & Biomolecular Engineering, the University of Akron, Akron, OH 44325, USA.
| | - Ruth Nussinov
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA.
- Sackler Institute of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Buyong Ma
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA.
| |
Collapse
|
321
|
β-barrel Oligomers as Common Intermediates of Peptides Self-Assembling into Cross-β Aggregates. Sci Rep 2018; 8:10353. [PMID: 29985420 PMCID: PMC6037789 DOI: 10.1038/s41598-018-28649-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/22/2018] [Indexed: 12/22/2022] Open
Abstract
Oligomers populated during the early amyloid aggregation process are more toxic than mature fibrils, but pinpointing the exact toxic species among highly dynamic and heterogeneous aggregation intermediates remains a major challenge. β-barrel oligomers, structurally-determined recently for a slow-aggregating peptide derived from αB crystallin, are attractive candidates for exerting amyloid toxicity due to their well-defined structures as therapeutic targets and compatibility to the "amyloid-pore" hypothesis of toxicity. To assess whether β-barrel oligomers are common intermediates to amyloid peptides - a necessary step toward associating β-barrel oligomers with general amyloid cytotoxicity, we computationally studied the oligomerization and fibrillization dynamics of seven well-studied fragments of amyloidogenic proteins with different experimentally-determined aggregation morphologies and cytotoxicity. In our molecular dynamics simulations, β-barrel oligomers were only observed in five peptides self-assembling into the characteristic cross-β aggregates, but not the other two that formed polymorphic β-rich aggregates as reported experimentally. Interestingly, the latter two peptides were previously found nontoxic. Hence, the observed correlation between β-barrel oligomers formation and cytotoxicity supports the hypothesis of β-barrel oligomers as the common toxic intermediates of amyloid aggregation.
Collapse
|
322
|
Röder K, Wales DJ. Predicting Pathways between Distant Configurations for Biomolecules. J Chem Theory Comput 2018; 14:4271-4278. [DOI: 10.1021/acs.jctc.8b00370] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Konstantin Röder
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - David J. Wales
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| |
Collapse
|
323
|
Kouza M, Co NT, Li MS, Kmiecik S, Kolinski A, Kloczkowski A, Buhimschi IA. Kinetics and mechanical stability of the fibril state control fibril formation time of polypeptide chains: A computational study. J Chem Phys 2018; 148:215106. [PMID: 29884031 DOI: 10.1063/1.5028575] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Fibril formation resulting from protein misfolding and aggregation is a hallmark of several neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. Despite much progress in the understanding of the protein aggregation process, the factors governing fibril formation rates and fibril stability have not been fully understood. Using lattice models, we have shown that the fibril formation time is controlled by the kinetic stability of the fibril state but not by its energy. Having performed all-atom explicit solvent molecular dynamics simulations with the GROMOS43a1 force field for full-length amyloid beta peptides Aβ40 and Aβ42 and truncated peptides, we demonstrated that kinetic stability can be accessed via mechanical stability in such a way that the higher the mechanical stability or the kinetic stability, the faster the fibril formation. This result opens up a new way for predicting fibril formation rates based on mechanical stability that may be easily estimated by steered molecular dynamics.
Collapse
Affiliation(s)
- Maksim Kouza
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Nguyen Truong Co
- Institute of Physics, Polish Academy of Sciences, Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Mai Suan Li
- Institute of Physics, Polish Academy of Sciences, Lotnikow 32/46, 02-668 Warsaw, Poland
| | - Sebastian Kmiecik
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Andrzej Kolinski
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Andrzej Kloczkowski
- Battelle Center for Mathematical Medicine, The Research Nationwide Children's Hospital, 575 Children's Crossroad, Columbus, Ohio 43215, USA
| | | |
Collapse
|
324
|
Chu T, Shu Y, Qu Y, Gao S, Zhang L. miR-26b inhibits total neurite outgrowth, promotes cells apoptosis and downregulates neprilysin in Alzheimer's disease. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3383-3390. [PMID: 31949715 PMCID: PMC6962878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/14/2018] [Indexed: 06/10/2023]
Abstract
This study aimed to investigate the effect of miR-26b expression on neurites outgrowth and cells apoptosis in PC12 cellular model of Alzheimer's disease (AD). PC12 cells were stimulated by nerve growth factor and insulted by Aβ1-42 to establish PC12 cellular AD model. Methyl thiazolyl tetrazolium (MTT) assay was then used to detect cells viability. Blank mimic, miR-26b mimic, blank inhibitor and miR-26b inhibitor plasmids were transferred into PC12 cellular AD models as NC1-mimic, miR-26b mimic, NC2-inhibitor and miR-26b inhibitor groups respectively. mRNA level, protein level, total neurite outgrowth and cells apoptosis were determined by qPCR, western blot, microscope and Hoechst/PI, respectively. MTT reduction rate was decreased in Aβ1-42 insult group compared to control group (P<0.001). After plasmids transfection, the total neuritis growth was found to be reduced in miR-26b mimic group compared with NC1-mimic group (P<0.05) while was elevated in miR-26b inhibitor group compared with NC2-inhibitor group (P<0.01). As to cells apoptosis, the percentage of apoptosis cells was increased in miR-26b mimic group than NC1-mimic group (P<0.05), and was decreased in miR-26b inhibitor group than NC2-inhibitor group (P<0.05). In addition, neprilysin (NEP) protein and mRNA expressions were decreased in miR-26b mimic group than NC1-mimic group and was increased in miR-26b inhibitor group than NC2-inhibitor group. However, protein or mRNA expression of EIF2S1 and αTTP was not affected by miR-26b. In conclusion, miR-26b inhibits neurite outgrowth, induces cells apoptosis and downregulates NEP expression in PC12 cellular AD model.
Collapse
Affiliation(s)
- Tingting Chu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical UniversityHarbin, China
- The First Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Yongwei Shu
- The First Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Yang Qu
- The First Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical UniversityHarbin, China
| | - Shasha Gao
- Medical Department, Peide HospitalMishan, China
| | - Liming Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical UniversityHarbin, China
| |
Collapse
|
325
|
Zhang T, Pauly T, Nagel-Steger L. Stoichiometric Zn2+ interferes with the self-association of Aβ42: Insights from size distribution analysis. Int J Biol Macromol 2018; 113:631-639. [DOI: 10.1016/j.ijbiomac.2018.02.123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/16/2018] [Accepted: 02/19/2018] [Indexed: 12/17/2022]
|
326
|
Xi W, Hansmann UHE. Conversion between parallel and antiparallel β-sheets in wild-type and Iowa mutant Aβ 40 fibrils. J Chem Phys 2018; 148:045103. [PMID: 29390821 DOI: 10.1063/1.5016166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Using a variant of Hamilton-replica-exchange, we study for wild type and Iowa mutant Aβ40 the conversion between fibrils with antiparallel β-sheets and such with parallel β-sheets. We show that wild type and mutant form distinct salt bridges that in turn stabilize different fibril organizations. The conversion between the two fibril forms leads to the release of small aggregates that in the Iowa mutant may shift the equilibrium from fibrils to more toxic oligomers.
Collapse
Affiliation(s)
- Wenhui Xi
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Ulrich H E Hansmann
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| |
Collapse
|
327
|
Katyal N, Agarwal M, Sen R, Kumar V, Deep S. Paradoxical Effect of Trehalose on the Aggregation of α-Synuclein: Expedites Onset of Aggregation yet Reduces Fibril Load. ACS Chem Neurosci 2018; 9:1477-1491. [PMID: 29601727 DOI: 10.1021/acschemneuro.8b00056] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Aggregation of α-synuclein is closely connected to the pathology of Parkinson's disease. The phenomenon involves multiple steps, commenced by partial misfolding and eventually leading to mature amyloid fibril formation. Trehalose, a widely accepted osmolyte, has been shown previously to inhibit aggregation of various globular proteins owing to its ability to prevent the initial unfolding of protein. In this study, we have examined if it behaves in a similar fashion with intrinsically disordered protein α-synuclein and possesses the potential to act as therapeutic agent against Parkinson's disease. It was observed experimentally that samples coincubated with trehalose fibrillate faster compared to the case in its absence. Molecular dynamics simulations suggested that this initial acceleration is manifestation of trehalose's tendency to perturb the conformational transitions between different conformers of monomeric protein. It stabilizes the aggregation prone "extended" conformer of α-synuclein, by binding to its exposed acidic residues of the C terminus. It also favors the β-rich oligomers once formed. Interestingly, the total fibrils formed are still promisingly less since it accelerates the competing pathway toward formation of amorphous aggregates.
Collapse
Affiliation(s)
- Nidhi Katyal
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| | - Manish Agarwal
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| | - Raktim Sen
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| | - Vinay Kumar
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| | - Shashank Deep
- Department of Chemistry, Indian Institute of Technology, Delhi, Hauz-Khas, New Delhi 110016, India
| |
Collapse
|
328
|
Nguyen PH, del Castillo-Frias MP, Berthoumieux O, Faller P, Doig AJ, Derreumaux P. Amyloid-β/Drug Interactions from Computer Simulations and Cell-Based Assays. J Alzheimers Dis 2018; 64:S659-S672. [DOI: 10.3233/jad-179902] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Phuong H. Nguyen
- Laboratoire de Biochimie Théorique, UPR 9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, IBPC, Paris, France
| | - Maria P. del Castillo-Frias
- Manchester Institute of Biotechnology and Department of Chemistry, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Olivia Berthoumieux
- CNRS, LCC (Laboratoire de Chimie de Coordination), Toulouse Cedex 4, France et Université de Toulouse, UPS, INPT, Toulouse Cedex 4, France
| | - Peter Faller
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR7177), Université de Strasbourg, Strasbourg, France
| | - Andrew J. Doig
- Manchester Institute of Biotechnology and Department of Chemistry, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Philippe Derreumaux
- Laboratoire de Biochimie Théorique, UPR 9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, IBPC, Paris, France
| |
Collapse
|
329
|
Ngo ST, Hung HM, Hong ND, Tung NT. The influences of E22Q mutant on solvated 3Aβ 11-40 peptide: A REMD study. J Mol Graph Model 2018; 83:122-128. [PMID: 29902674 DOI: 10.1016/j.jmgm.2018.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/03/2018] [Accepted: 06/01/2018] [Indexed: 10/14/2022]
Abstract
The residue E22 plays a critical role in the aggregation process of Amyloid beta (Aβ) peptides. The effect of E22Q mutant on the shapes of the solvated Aβ11-40 trimer is clarified using a replica exchange molecular dynamics (REMD) simulation employing ∼20.6 μs of MD simulations with 48 disparate replicas. The increase of intramolecular polar contacts and salt bridge between the residue D23 to residues (24-29) was observed. The residual secondary structure of the mutated trimer is shifted in a similar way to the picture observed in previous investigations of F19W mutant. The free energy surface (FES) of the mutated E22Q system has a fewer number of minima in comparison with the wild-type trimer. The optimized shapes of the mutated E22Q form a significant increase in beta structure (47%) and serious decrease in coil content (46%) compared with the wild-type (of 36 and 56%, respectively). The binding affinity of constituting chains to the rest is of -43.7 ± 6.5 kcal/mol, implying that the representative structure of E22Q is more stable than the wild-type one. Furthermore, the E22Q mutant increases the size of stable structures due to larger collision cross section (CCS) and solvent accessible area (SASA). The observed results may enhance the Aβ inhibition throughout the contribution to the knowledge of the Aβ oligomerization/aggregation.
Collapse
Affiliation(s)
- Son Tung Ngo
- Computational Chemistry Research Group, Ton Duc Thang University, Ho Chi Minh City, Vietnam; Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam.
| | - Huynh Minh Hung
- Department of Chemistry, KU Leuven, Celestijnenlaan 200F, B-3001, Leuven, Belgium
| | - Nam Dao Hong
- University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Nguyen Thanh Tung
- Institute of Materials Science and Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Vietnam.
| |
Collapse
|
330
|
Reuter B, Weber M, Fackeldey K, Röblitz S, Garcia ME. Generalized Markov State Modeling Method for Nonequilibrium Biomolecular Dynamics: Exemplified on Amyloid β Conformational Dynamics Driven by an Oscillating Electric Field. J Chem Theory Comput 2018; 14:3579-3594. [DOI: 10.1021/acs.jctc.8b00079] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Bernhard Reuter
- University of Kassel, Institute of Physics, Theoretical Physics II, Heinrich-Plett-Str. 40, 34132 Kassel, Germany
- Zuse Institute Berlin (ZIB), Takustraße 7, 14195 Berlin, Germany
| | - Marcus Weber
- Zuse Institute Berlin (ZIB), Takustraße 7, 14195 Berlin, Germany
| | - Konstantin Fackeldey
- Zuse Institute Berlin (ZIB), Takustraße 7, 14195 Berlin, Germany
- Institute of Mathematics, Technical University Berlin, Straße des 17. Juni 136, 10623 Berlin, Germany
| | - Susanna Röblitz
- Zuse Institute Berlin (ZIB), Takustraße 7, 14195 Berlin, Germany
| | - Martin E. Garcia
- University of Kassel, Institute of Physics, Theoretical Physics II, Heinrich-Plett-Str. 40, 34132 Kassel, Germany
| |
Collapse
|
331
|
Frederix PWJM, Patmanidis I, Marrink SJ. Molecular simulations of self-assembling bio-inspired supramolecular systems and their connection to experiments. Chem Soc Rev 2018; 47:3470-3489. [PMID: 29688238 PMCID: PMC5961611 DOI: 10.1039/c8cs00040a] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Indexed: 01/01/2023]
Abstract
In bionanotechnology, the field of creating functional materials consisting of bio-inspired molecules, the function and shape of a nanostructure only appear through the assembly of many small molecules together. The large number of building blocks required to define a nanostructure combined with the many degrees of freedom in packing small molecules has long precluded molecular simulations, but recent advances in computational hardware as well as software have made classical simulations available to this strongly expanding field. Here, we review the state of the art in simulations of self-assembling bio-inspired supramolecular systems. We will first discuss progress in force fields, simulation protocols and enhanced sampling techniques using recent examples. Secondly, we will focus on efforts to enable the comparison of experimentally accessible observables and computational results. Experimental quantities that can be measured by microscopy, spectroscopy and scattering can be linked to simulation output either directly or indirectly, via quantum mechanical or semi-empirical techniques. Overall, we aim to provide an overview of the various computational approaches to understand not only the molecular architecture of nanostructures, but also the mechanism of their formation.
Collapse
Affiliation(s)
- Pim W. J. M. Frederix
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials , University of Groningen , Groningen , The Netherlands . ;
| | - Ilias Patmanidis
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials , University of Groningen , Groningen , The Netherlands . ;
| | - Siewert J. Marrink
- Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials , University of Groningen , Groningen , The Netherlands . ;
| |
Collapse
|
332
|
Ren B, Liu Y, Zhang Y, Cai Y, Gong X, Chang Y, Xu L, Zheng J. Genistein: A Dual Inhibitor of Both Amyloid β and Human Islet Amylin Peptides. ACS Chem Neurosci 2018; 9:1215-1224. [PMID: 29432676 DOI: 10.1021/acschemneuro.8b00039] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abnormal misfolding and aggregation of amyloid peptides into amyloid fibrils are common and critical pathological events in many neurodegenerative diseases. Most inhibitors or drugs have been developed to prevent amyloid aggregation of a specific peptide, showing sequence-dependent inhibition mechanisms. It is more challenging to develop or discover inhibitors capable of preventing the aggregation of two or more different amyloid peptides. Genistein, a major phytoestrogen in soybean, has been widely used as an anti-inflammation and cerebrovascular drug due to its antioxidation and antiacetylcholinesterase effects. Herein, we examine the inhibitory effects of genistein on the aggregation of amyloid-β (Aβ, associated with Alzheimer's disease) and human islet amylin (hIAPP, associated with type 2 diabetes) and Aβ- and hIAPP-induced neurotoxicity using a combination of experimental and computational approaches. Collective experimental results from thioflavin T (ThT), atomic force microscopy (AFM), and circular dichroism (CD) demonstrate that genistein shows strong inhibition ability to prevent the conformational transition of both Aβ and hIAPP monomers to β-sheet structures, thus reducing final amyloid fibrillization from Aβ and hIAPP monomer aggregation by 40-63%. Further 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT), lactate dehydrogenase (LDH), and large unilamellar vesicle (LUV) assays show that genistein helps to increase cell viability, decrease cell apoptosis, and reduce cell membrane leakage, where the cell protection effect of genistein is likely correlated with its reduced membrane leakage. Comparative molecular dynamics (MD) simulations reveal that genistein prefers to bind the β-sheet groove, a common structural motif of amyloid fibrils, of both Aβ and hIAPP oligomers to interfere with their self-aggregation. This work for the first time demonstrates genistein as a dual inhibitor of Aβ and hIAPP aggregation. Further structural optimization and refinement of genistein may generate a series of effective sequence-independent inhibitors against the aggregation and toxicity of different amyloid peptides.
Collapse
Affiliation(s)
- Baiping Ren
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, P. R. China
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Yonglan Liu
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Yanxian Zhang
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Yongqing Cai
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Xiong Gong
- Department of Polymer Engineering, College of Polymer Science and Polymer Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Yung Chang
- R&D Center for Membrane Technology and Department of Chemical Engineering, Chung Yuan Christian University, Chung-Li, Taoyuan 320, Taiwan
| | - Lijian Xu
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, P. R. China
| | - Jie Zheng
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| |
Collapse
|
333
|
Wang X, Wang X, Guo Z. Metal-involved theranostics: An emerging strategy for fighting Alzheimer’s disease. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2018.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
334
|
van der Munnik NP, Sajib MSJ, Moss MA, Wei T, Uline MJ. Determining the Potential of Mean Force for Amyloid-β Dimerization: Combining Self-Consistent Field Theory with Molecular Dynamics Simulation. J Chem Theory Comput 2018; 14:2696-2704. [PMID: 29562134 DOI: 10.1021/acs.jctc.7b01057] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Amyloid-β (Aβ) protein aggregates through a complex pathway to progress from monomers to soluble oligomers and ultimately insoluble fibrils. Because of the dynamic nature of aggregation, it has proven exceedingly difficult to determine the precise interactions that lead to the formation of transient oligomers. Here, a statistical thermodynamic model has been developed to elucidate these interactions. Aβ1-42 was simulated using fully atomistic replica exchange molecular dynamics. We use an ensemble of approximately 5 × 105 configurations taken from simulation as input in a self-consistent field theory that explicitly accounts for the size, shape, and charge distribution of both the amino acids comprising Aβ and all molecular species present in solution. The solution of the model equations provides a prediction of the probabilities of the configurations of the Aβ dimer and the potential of mean force between two monomers during the dimerization process. This model constitutes a reliable methodology to elucidate the underlying physics of the Aβ dimerization process as a function of pH, temperature, and salt concentration. The results obtained with this new model could be valuable in the design of Aβ oligomerization inhibitors, a prospective therapeutic for Alzheimer's disease.
Collapse
Affiliation(s)
- Nicholas P van der Munnik
- Department of Chemical Engineering , University of South Carolina , Columbia , South Carolina 29208 , United States
| | - Md Symon Jahan Sajib
- Department of Chemical Engineering , Lamar University , Beaumont , Texas 77705 , United States
| | - Melissa A Moss
- Department of Chemical Engineering , University of South Carolina , Columbia , South Carolina 29208 , United States.,Biomedical Engineering Program , University of South Carolina , Columbia , South Carolina 29208 , United States
| | - Tao Wei
- Department of Chemical Engineering , Lamar University , Beaumont , Texas 77705 , United States.,Department of Chemical Engineering , Howard University , Washington D.C. , 20059 , United States
| | - Mark J Uline
- Department of Chemical Engineering , University of South Carolina , Columbia , South Carolina 29208 , United States.,Biomedical Engineering Program , University of South Carolina , Columbia , South Carolina 29208 , United States
| |
Collapse
|
335
|
Owen MC, Kulig W, Poojari C, Rog T, Strodel B. Physiologically-relevant levels of sphingomyelin, but not GM1, induces a β-sheet-rich structure in the amyloid-β(1-42) monomer. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1709-1720. [PMID: 29626441 DOI: 10.1016/j.bbamem.2018.03.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/29/2018] [Accepted: 03/31/2018] [Indexed: 01/07/2023]
Abstract
To resolve the contribution of ceramide-containing lipids to the aggregation of the amyloid-β protein into β-sheet rich toxic oligomers, we employed molecular dynamics simulations to study the effect of cholesterol-containing bilayers comprised of POPC (70% POPC, and 30% cholesterol) and physiologically relevant concentrations of sphingomyelin (SM) (30% SM, 40% POPC, and 30% cholesterol), and the GM1 ganglioside (5% GM1, 70% POPC, and 25% cholesterol). The increased bilayer rigidity provided by SM (and to a lesser degree, GM1) reduced the interactions between the SM-enriched bilayer and the N-terminus of Aβ42 (and also residues Ser26, Asn27, and Lys28), which facilitated the formation of a β-sheet in the normally disordered N-terminal region. Aβ42 remained anchored to the SM-enriched bilayer through hydrogen bonds with the side chain of Arg5. With β-sheets in the at the N and C termini, the structure of Aβ42 in the sphingomyelin-enriched bilayer most resembles β-sheet-rich structures found in higher-ordered Aβ fibrils. Conversely, when bound to a bilayer comprised of 5% GM1, the conformation remained similar to that observed in the absence of GM1, with Aβ42 only making contact with one or two GM1 molecules. This article is part of a Special Issue entitled: Protein Aggregation and Misfolding at the Cell Membrane Interface edited by Ayyalusamy Ramamoorthy.
Collapse
Affiliation(s)
- Michael C Owen
- Institute of Complex Systems: Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany; CEITEC - Central European Institute of Technology, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic.
| | - Waldemar Kulig
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland; Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| | - Chetan Poojari
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland; Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| | - Tomasz Rog
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland; Department of Physics, Tampere University of Technology, P.O. Box 692, FI-33101 Tampere, Finland
| | - Birgit Strodel
- Institute of Complex Systems: Structural Biochemistry (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany; Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
336
|
Cheignon C, Tomas M, Bonnefont-Rousselot D, Faller P, Hureau C, Collin F. Oxidative stress and the amyloid beta peptide in Alzheimer's disease. Redox Biol 2018; 14:450-464. [PMID: 29080524 PMCID: PMC5680523 DOI: 10.1016/j.redox.2017.10.014] [Citation(s) in RCA: 1443] [Impact Index Per Article: 206.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/14/2017] [Accepted: 10/17/2017] [Indexed: 01/12/2023] Open
Abstract
Oxidative stress is known to play an important role in the pathogenesis of a number of diseases. In particular, it is linked to the etiology of Alzheimer's disease (AD), an age-related neurodegenerative disease and the most common cause of dementia in the elderly. Histopathological hallmarks of AD are intracellular neurofibrillary tangles and extracellular formation of senile plaques composed of the amyloid-beta peptide (Aβ) in aggregated form along with metal-ions such as copper, iron or zinc. Redox active metal ions, as for example copper, can catalyze the production of Reactive Oxygen Species (ROS) when bound to the amyloid-β (Aβ). The ROS thus produced, in particular the hydroxyl radical which is the most reactive one, may contribute to oxidative damage on both the Aβ peptide itself and on surrounding molecule (proteins, lipids, …). This review highlights the existing link between oxidative stress and AD, and the consequences towards the Aβ peptide and surrounding molecules in terms of oxidative damage. In addition, the implication of metal ions in AD, their interaction with the Aβ peptide and redox properties leading to ROS production are discussed, along with both in vitro and in vivo oxidation of the Aβ peptide, at the molecular level.
Collapse
Affiliation(s)
- C Cheignon
- LCC (Laboratoire de Chimie de Coordination), CNRS UPR 8241, 205 route de Narbonne, 31062 Toulouse Cedex 09, France; Université de Toulouse; UPS, INPT, 31077 Toulouse, France
| | - M Tomas
- LCC (Laboratoire de Chimie de Coordination), CNRS UPR 8241, 205 route de Narbonne, 31062 Toulouse Cedex 09, France; Université de Toulouse; UPS, INPT, 31077 Toulouse, France
| | - D Bonnefont-Rousselot
- Department of Metabolic Biochemistry, La Pitié Salpêtrière-Charles Foix University Hospital (AP-HP), Paris, France; Department of Biochemistry, Faculty of Pharmacy, Paris Descartes University, Paris, France; CNRS UMR8258 - INSERM U1022, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | - P Faller
- Biometals and Biology Chemistry, Institut de Chimie (CNRS UMR 7177), University of Strasbourg, 4 rue B. Pascal, 67081 Strasbourg Cedex, France
| | - C Hureau
- LCC (Laboratoire de Chimie de Coordination), CNRS UPR 8241, 205 route de Narbonne, 31062 Toulouse Cedex 09, France; Université de Toulouse; UPS, INPT, 31077 Toulouse, France
| | - F Collin
- LCC (Laboratoire de Chimie de Coordination), CNRS UPR 8241, 205 route de Narbonne, 31062 Toulouse Cedex 09, France; Université de Toulouse; UPS, INPT, 31077 Toulouse, France.
| |
Collapse
|
337
|
Dong X, Qiao Q, Qian Z, Wei G. Recent computational studies of membrane interaction and disruption of human islet amyloid polypeptide: Monomers, oligomers and protofibrils. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018. [PMID: 29530482 DOI: 10.1016/j.bbamem.2018.03.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The amyloid deposits of human islet amyloid polypeptide (hIAPP) are found in type 2 diabetes patients. hIAPP monomer is intrinsically disordered in solution, whereas it can form amyloid fibrils both in vivo and in vitro. Extensive evidence suggests that hIAPP causes the disruption of cellular membrane, and further induces cytotoxicity and the death of islet β-cells in pancreas. The presence of membrane also accelerates the hIAPP fibril formation. hIAPP oligomers and protofibrils in the early stage of aggregation were reported to be the most cytotoxic, disrupting the membrane integrity and giving rise to the pathological process. The detailed molecular mechanisms of hIAPP-membrane interactions and membrane disruption are complex and remain mostly unknown. Here in this review, we focus on recent computational studies that investigated the interactions of full length and fragmentary hIAPP monomers, oligomers and protofibrils with anionic, zwitterionic and mixed anionic-zwitterionic lipid bilayers. We mainly discuss the binding orientation of monomers at membrane surface, the conformational ensemble and the oligomerization of hIAPP inside membranes, the effect of lipid composition on hIAPP oligomers/protofibrils-membrane interactions, and the hIAPP-induced membrane perturbation. This review provides mechanistic insights into the interactions between hIAPP and lipid bilayers with different lipid composition at an atomistic level, which is helpful to understand the hIAPP cytotoxicity mediated by membrane. This article is part of a Special Issue entitled: Protein Aggregation and Misfolding at the Cell Membrane Interface edited by Ayyalusamy Ramamoorthy.
Collapse
Affiliation(s)
- Xuewei Dong
- Department of Physics, State Key Laboratory of Surface Physics, Key Laboratory for Computational Physical Science (Ministry of Education), Collaborative Innovation Center of Advanced Microstructures (Nanjing), Fudan University, Shanghai 200433, China
| | - Qin Qiao
- Digital Medical Research Center, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Shanghai Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention, Shanghai 200032, China.
| | - Zhenyu Qian
- Key Laboratory of Exercise and Health Sciences (Ministry of Education) and School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Guanghong Wei
- Department of Physics, State Key Laboratory of Surface Physics, Key Laboratory for Computational Physical Science (Ministry of Education), Collaborative Innovation Center of Advanced Microstructures (Nanjing), Fudan University, Shanghai 200433, China.
| |
Collapse
|
338
|
Abstract
The aggregation of the Aβ peptide (Aβ1-42) to form fibrils is a key feature of Alzheimer's disease. The mechanism is thought to be a nucleation stage followed by an elongation process. The elongation stage involves the consecutive addition of monomers to one end of the growing fibril. The aggregation process proceeds in a stop-and-go fashion and may involve off-pathway aggregates, complicating experimental and computational studies. Here we present exploration of a well-defined region in the free and potential energy landscapes for the Aβ17-42 pentamer. We find that the ideal aggregation process agrees with the previously reported dock-lock mechanism. We also analyze a large number of additional stable structures located on the multifunnel energy landscape, which constitute kinetic traps. The key contributors to the formation of such traps are misaligned strong interactions, for example the stacking of F19 and F20, as well as entropic contributions. Our results suggest that folding templates for aggregation are a necessity and that aggregation studies could employ such species to obtain a more detailed description of the process.
Collapse
Affiliation(s)
- Konstantin Röder
- Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge , CB2 1EW , United Kingdom
| | - David J Wales
- Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge , CB2 1EW , United Kingdom
| |
Collapse
|
339
|
Ligand field molecular dynamics simulation of Pt(II)-phenanthroline binding to N-terminal fragment of amyloid-β peptide. PLoS One 2018; 13:e0193668. [PMID: 29509784 PMCID: PMC5839559 DOI: 10.1371/journal.pone.0193668] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/15/2018] [Indexed: 12/15/2022] Open
Abstract
We report microsecond timescale molecular dynamics simulation of the complex formed between Pt(II)-phenanthroline and the 16 N-terminal residues of the Aβ peptide that is implicated in the onset of Alzheimer’s disease, along with equivalent simulations of the metal-free peptide. Simulations from a variety of starting points reach equilibrium within 100 ns, as judged by root mean square deviation and radius of gyration. Platinum-bound peptides deviate rather more from starting points, and adopt structures with larger radius of gyration, than their metal-free counterparts. Residues bound directly to Pt show smaller fluctuation, but others actually move more in the Pt-bound peptide. Hydrogen bonding within the peptide is disrupted by binding of Pt, whereas the presence of salt-bridges are enhanced.
Collapse
|
340
|
Ginex T, Trius M, Luque FJ. Computational Study of the Aza-Michael Addition of the Flavonoid (+)-Taxifolin in the Inhibition of β-Amyloid Fibril Aggregation. Chemistry 2018; 24:5813-5824. [PMID: 29384229 DOI: 10.1002/chem.201706072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Indexed: 01/30/2023]
Abstract
Inhibition of abnormal protein self-aggregation is an attractive strategy against amyloidogenic diseases, but has found limited success due to the complexity of protein self-assembly, the absence of fully reproducible aggregation assays, and the scarce knowledge of the inhibition mechanisms by small molecules. In this context, catechol-containing compounds may lead to covalent adducts with amyloid fibrils that interfere with the aggregation process. In particular, the covalent adduct formed between the oxidized form of (+)-taxifolin and an β-amyloid (Aβ42) suggests the involvement of a specific recognition motif that enables the chemical reaction with Aβ42. In this study, we have examined the mechanisms implicated in the aza-Michael addition of the o-quinone species of (+)-taxifolin with Aβ42 fibrils. The results support the binding of (+)-taxifolin to the hydrophobic groove delimited by the edges defined by Lys16 and Glu22 residues in the fibril. The chemical reaction proceeds through the nucleophilic attack of the deprotonated amino group of a Lys16 residue in a process activated by the interaction between the o-quinone ring with a vicinal Lys16 residue, as well as by a water-assisted proton transfer, which is the rate-limiting step of the reaction. This specific inhibition mechanism, which may explain the enhanced anti-aggregating activity of oxidized flavonoids compared to fresh compounds, holds promise for developing disease-modifying therapies.
Collapse
Affiliation(s)
- Tiziana Ginex
- Department of Nutrition, Food Science, and Gastronomy, Faculty of Pharmacy and Institute of Biomedicine, Campus Torribera, University of Barcelona, Santa Coloma de Gramenet, 08921, Spain
| | - Marta Trius
- Department of Nutrition, Food Science, and Gastronomy, Faculty of Pharmacy and Institute of Biomedicine, Campus Torribera, University of Barcelona, Santa Coloma de Gramenet, 08921, Spain
| | - F Javier Luque
- Department of Nutrition, Food Science, and Gastronomy, Faculty of Pharmacy and Institute of Biomedicine, Campus Torribera, University of Barcelona, Santa Coloma de Gramenet, 08921, Spain
| |
Collapse
|
341
|
Amyloid growth and membrane damage: Current themes and emerging perspectives from theory and experiments on Aβ and hIAPP. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1625-1638. [PMID: 29501606 DOI: 10.1016/j.bbamem.2018.02.022] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 12/15/2022]
Abstract
Alzheimer's Disease (AD) and Type 2 diabetes mellitus (T2DM) are two incurable diseases both hallmarked by an abnormal deposition of the amyloidogenic peptides Aβ and Islet Amyloid Polypeptide (IAPP) in affected tissues. Epidemiological data demonstrate that patients suffering from diabetes are at high risk of developing AD, thus making the search for factors common to the two pathologies of special interest for the design of new therapies. Accumulating evidence suggests that the toxic properties of both Aβ or IAPP are ascribable to their ability to damage the cell membrane. However, the molecular details describing Aβ or IAPP interaction with membranes are poorly understood. This review focuses on biophysical and in silico studies addressing these topics. Effects of calcium, cholesterol and membrane lipid composition in driving aberrant Aβ or IAPP interaction with the membrane will be specifically considered. The cross correlation of all these factors appears to be a key issue not only to shed light in the countless and often controversial reports relative to this area but also to gain valuable insights into the central events leading to membrane damage caused by amyloidogenic peptides. This article is part of a Special Issue entitled: Protein Aggregation and Misfolding at the Cell Membrane Interface edited by Ayyalusamy Ramamoorthy.
Collapse
|
342
|
Jana AK, Batkulwar KB, Kulkarni MJ, Sengupta N. Glycation induces conformational changes in the amyloid-β peptide and enhances its aggregation propensity: molecular insights. Phys Chem Chem Phys 2018; 18:31446-31458. [PMID: 27827482 DOI: 10.1039/c6cp05041g] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The cytotoxicity of the amyloid beta (Aβ) peptide, implicated in the pathogenesis of Alzheimer's disease (AD), can be enhanced by its post-translational glycation, a series of non-enzymatic reactions with reducing sugars and reactive dicarbonyls. However, little is known about the underlying mechanisms that potentially enhance the cytotoxicity of the advanced glycation modified Aβ. In this work, fully atomistic molecular dynamics (MD) simulations are exploited to obtain direct molecular insights into the process of early Aβ self-assembly in the presence and absence of glycated lysine residues. Analyses of data exceeding cumulative timescales of 1 microsecond for each system reveal that glycation results in a stronger enthalpy of association between Aβ monomers and lower conformational entropy, in addition to a sharp overall increase in the beta-sheet content. Further analyses reveal that the enhanced interactions originate, in large part, due to markedly stronger, as well as new, inter-monomer salt bridging propensities in the glycated variety. Interestingly, these conformational and energetic effects are broadly reflected in preformed protofibrillar forms of Aβ small oligomers modified with glycation. Our combined results imply that glycation consolidates Aβ self-assembly regardless of its point of occurrence in the pathway. They provide a basis for further mechanistic studies and therapeutic endeavors that could potentially result in novel ways of combating AGE related AD progression.
Collapse
Affiliation(s)
- Asis K Jana
- Physical Chemistry Division, CSIR-National Chemical Laboratory, Pune 411008, India and Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Kedar B Batkulwar
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India and Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India.
| | - Mahesh J Kulkarni
- Academy of Scientific and Innovative Research (AcSIR), New Delhi, India and Biochemical Sciences Division, CSIR-National Chemical Laboratory, Pune 411008, India.
| | - Neelanjana Sengupta
- Dept. of Biological Sciences, Indian Institute of Science Education and Research (IISER) Kolkata, Mohanpur 741 246, W. Bengal, India.
| |
Collapse
|
343
|
Hane FT, Lee BY, Leonenko Z. Recent Progress in Alzheimer's Disease Research, Part 1: Pathology. J Alzheimers Dis 2018; 57:1-28. [PMID: 28222507 DOI: 10.3233/jad-160882] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The field of Alzheimer's disease (AD) research has grown exponentially over the past few decades, especially since the isolation and identification of amyloid-β from postmortem examination of the brains of AD patients. Recently, the Journal of Alzheimer's Disease (JAD) put forth approximately 300 research reports which were deemed to be the most influential research reports in the field of AD since 2010. JAD readers were asked to vote on these most influential reports. In this 3-part review, we review the results of the 300 most influential AD research reports to provide JAD readers with a readily accessible, yet comprehensive review of the state of contemporary research. Notably, this multi-part review identifies the "hottest" fields of AD research providing guidance for both senior investigators as well as investigators new to the field on what is the most pressing fields within AD research. Part 1 of this review covers pathogenesis, both on a molecular and macro scale. Part 2 review genetics and epidemiology, and part 3 covers diagnosis and treatment. This part of the review, pathology, reviews amyloid-β, tau, prions, brain structure, and functional changes with AD and the neuroimmune response of AD.
Collapse
Affiliation(s)
- Francis T Hane
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.,Department of Chemistry, Lakehead University, Thunder Bay, ON, Canada
| | - Brenda Y Lee
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Zoya Leonenko
- Department of Biology, University of Waterloo, Waterloo, ON, Canada.,Department of Physics and Astronomy, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
344
|
Baweja L, Roche J. Pushing the Limits of Structure-Based Models: Prediction of Nonglobular Protein Folding and Fibrils Formation with Go-Model Simulations. J Phys Chem B 2018; 122:2525-2535. [DOI: 10.1021/acs.jpcb.7b12129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Lokesh Baweja
- Department of Biochemistry, Molecular Biology and Biophysics, Iowa State University, Ames, Iowa 50011, United States
| | - Julien Roche
- Department of Biochemistry, Molecular Biology and Biophysics, Iowa State University, Ames, Iowa 50011, United States
| |
Collapse
|
345
|
Ren B, Jiang B, Hu R, Zhang M, Chen H, Ma J, Sun Y, Jia L, Zheng J. HP-β-cyclodextrin as an inhibitor of amyloid-β aggregation and toxicity. Phys Chem Chem Phys 2018; 18:20476-85. [PMID: 27405335 DOI: 10.1039/c6cp03582e] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Amyloid deposits of misfolded amyloid-β protein (Aβ) on neuronal cells are a pathological hallmark of Alzheimer's disease (AD). Prevention of the abnormal Aβ aggregation has been considered as a promising therapeutic strategy for AD treatment. To prevent reinventing the wheel, we proposed to search the existing drug database for other diseases to identify potential Aβ inhibitors. Herein, we reported the inhibitory activity of HP-β-cyclodextrin (HP-β-CD), a well-known sugar used in drug delivery, genetic vector, environmental protection and treatment of Niemann-Pick disease type C1 (NPC1), against Aβ1-42 aggregation and Aβ-induced toxicity, with the aim of adding a new function as a sugar-based Aβ inhibitor. Experimental data showed that HP-β-CD molecules were not only nontoxic to cells, but also greatly inhibited Aβ fibrillization and reduced Aβ-induced toxicity in a concentration-dependent manner. At an optimal molar ratio of Aβ : HP-β-CD = 1 : 2, HP-β-CD enabled the reduction of 60% of Aβ fibrils and increased the cell viability to 92%. Such concentration-dependent inhibitor capacity of HP-β-CD was likely attributed to several combined effects, including the enhancement of Aβ-HP-β-CD interactions, prevention of structural transition of Aβ peptides towards β-sheet structures, and reduction of self-aggregation of HP-β-CD. In parallel, molecular simulations further revealed the atomic details of HP-β-CD interacting with the Aβ oligomer, showing that HP-β-CD had a high tendency to interact with hydrophobic residues of Aβ in two β-strands and the N-terminal tail. More importantly, we identified that the inner hydrophobic cavity of HP-β-CD was a key active site for Aβ inhibition. Once the inner cavity of HP-β-CD was blocked by a small hydrophobic molecule of ferulic acid, HP-β-CD completely lost its inhibition capacity against Aβ. Given the already established pharmaceutical functions of HP-β-CD in drug delivery, our findings suggest that HP-β-CD has great potential to be designed as a sugar-based Aβ inhibitor.
Collapse
Affiliation(s)
- Baiping Ren
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China and Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, USA.
| | - Binbo Jiang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China and Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, USA.
| | - Rundong Hu
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, USA.
| | - Mingzhen Zhang
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, USA.
| | - Hong Chen
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, USA.
| | - Jie Ma
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Tongji University, Shanghai 200092, China and Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, USA.
| | - 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
| | - Lingyun Jia
- School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, China
| | - Jie Zheng
- Department of Chemical & Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, USA.
| |
Collapse
|
346
|
Liu Y, Ren B, Zhang Y, Sun Y, Chang Y, Liang G, Xu L, Zheng J. Molecular simulation aspects of amyloid peptides at membrane interface. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1906-1916. [PMID: 29421626 DOI: 10.1016/j.bbamem.2018.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 12/13/2022]
Abstract
The interactions of amyloid peptides with cell membranes play an important role in maintaining the integrity and functionality of cell membrane. A thorough molecular-level understanding of the structure, dynamics, and interactions between amyloid peptides and cell membranes is critical to amyloid aggregation and toxicity mechanisms for the bench-to-bedside applications. Here we review the most recent computational studies of amyloid peptides at model cell membranes. Different mechanisms of action of amyloid peptides on/in cell membranes, targeted by different computational techniques at different lengthscales and timescales, are rationally discussed. Finally, we have proposed some new insights into the remaining challenges and perspectives for future studies to improve our understanding of the activity of amyloid peptides associated with protein-misfolding diseases. This article is part of a Special Issue entitled: Protein Aggregation and Misfolding at the Cell Membrane Interface edited by Ayyalusamy Ramamoorthy.
Collapse
Affiliation(s)
- Yonglan Liu
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, PR China; Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH 44325, United States
| | - Baiping Ren
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH 44325, United States
| | - Yanxian Zhang
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH 44325, United States
| | - 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
| | - Yung Chang
- R&D Center for Membrane Technology and Department of Chemical EngineeringChung Yuan Christian University, Chung-Li, Taoyuan 320, Taiwan
| | - Guizhao Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Lijian Xu
- Hunan Key Laboratory of Biomedical Nanomaterials and Devices, College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, PR China; Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH 44325, United States.
| | - Jie Zheng
- Department of Chemical and Biomolecular Engineering, The University of Akron, Akron, OH 44325, United States.
| |
Collapse
|
347
|
Ren H, Zhang Y, Guo S, Lin N, Deng L, Yue T, Huang F. Identifying Cu(ii)-amyloid peptide binding intermediates in the early stages of aggregation by resonance Raman spectroscopy: a simulation study. Phys Chem Chem Phys 2018; 19:31103-31112. [PMID: 29138762 DOI: 10.1039/c7cp06206k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aggregation of amyloid beta (Aβ) peptides plays a crucial role in the pathology and etiology of Alzheimer's disease. Experimental evidence shows that copper ion is an aggregation-prone species with the ability to coordinately bind to Aβ and further induce the formation of neurotoxic Aβ oligomers. However, the detailed structures of Cu(ii)-Aβ complexes have not been illustrated, and the kinetics and dynamics of the Cu(ii) binding are not well understood. Two Cu(ii)-Aβ complexes have been proposed to exist under physiological conditions, and another two might exist at higher pH values. By using ab initio simulations for the spontaneous resonance Raman and time domain stimulated resonance Raman spectroscopy signals, we obtained the characteristic Raman vibronic features of each complex. These signals contain rich structural information with high temporal resolution, enabling the characterization of transient states during the fast Cu-Aβ binding and interconversion processes.
Collapse
Affiliation(s)
- Hao Ren
- State Key Laboratory of Heavy Oil Processing, Center for Bioengineering & Biotechnology, China University of Petroleum (East China), Qingdao, 266580, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
348
|
Xi W, Vanderford EK, Hansmann UHE. Out-of-Register Aβ 42 Assemblies as Models for Neurotoxic Oligomers and Fibrils. J Chem Theory Comput 2018; 14:1099-1110. [PMID: 29357242 DOI: 10.1021/acs.jctc.7b01106] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We propose a variant of the recently found S-shaped Aβ1-42-motif that is characterized by out-of-register C-terminal β-strands. We show that chains with this structure can form not only fibrils that are compatible with the NMR signals but also barrel-shaped oligomers that resemble the ones formed by the much smaller cylindrin peptides. By running long all-atom molecular dynamics simulations at physiological temperatures with an explicit solvent, we study the stability of these constructs and show that they are plausible models for neurotoxic oligomers. After analyzing the transitions between different assemblies, we suggest a mechanism for amyloid formation in Alzheimer's disease.
Collapse
Affiliation(s)
- Wenhui Xi
- Department of Chemistry and Biochemistry, University of Oklahoma , Norman, Oklahoma 73019, United States
| | - Elliott K Vanderford
- Department of Chemistry and Biochemistry, University of Oklahoma , Norman, Oklahoma 73019, United States
| | - Ulrich H E Hansmann
- Department of Chemistry and Biochemistry, University of Oklahoma , Norman, Oklahoma 73019, United States
| |
Collapse
|
349
|
Lu Y, Shi XF, Salsbury FR, Derreumaux P. Influence of electric field on the amyloid-β(29-42) peptides embedded in a membrane bilayer. J Chem Phys 2018; 148:045105. [DOI: 10.1063/1.5018459] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Yan Lu
- School of Physics and Optoelectronic Engineering, Xidian University, Xi’an 710071, China
| | - Xiao-Feng Shi
- School of Physics and Optoelectronic Engineering, Xidian University, Xi’an 710071, China
| | - Freddie R. Salsbury
- Department of Physics, Wake Forest University, Winston-Salem, North Carolina 27106, USA
| | - Philippe Derreumaux
- Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique (IBPC), UPR9080 CNRS, Université Paris Diderot, Sorbonne Paris Cité, 13 Rue Pierre et Marie Curie, 75005 Paris, France
| |
Collapse
|
350
|
The Potential Protective Effect of Curcumin on Amyloid- β-42 Induced Cytotoxicity in HT-22 Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8134902. [PMID: 29568765 PMCID: PMC5820551 DOI: 10.1155/2018/8134902] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/09/2017] [Accepted: 11/26/2017] [Indexed: 12/23/2022]
Abstract
Background We aimed to investigate the effect and mechanism of curcumin (CUR) in Alzheimer's disease (AD). Methods Mouse hippocampal neuronal cell line HT-22 was treated with Aβ1–42 and/or CUR, and then cell viability was evaluated by cell counting kit 8, Beclin-l level was detected using western blotting, and the formation of autophagosomes was observed by transmission electron microscopy (TEM). Furthermore, transcriptome sequencing and analysis were performed in cells with Aβ1–42 alone or Aβ1–42 + CUR. Results Aβ1–42 treatment significantly inhibited cell viability compared with untreated cells (P < 0.01). After treatment for 48 h, CUR remarkably promoted cell viability compared with cell treated with Aβ1–42 alone (P < 0.01). Compared with cells treated with Aβ1–42 alone, the expression of Beclin-1 was slightly reduced in cells with combined treatment of Aβ1–42 with CUR (P < 0.05). Consistently, TEM results showed that CUR inhibited the formation of autophagosomes in cells treated with Aβ1–42. Furthermore, the protein-protein interaction network showed five key genes, including MYC, Cdh1, Acaca, Egr1, and CCnd1, likely involved in CUR effects. Conclusions CUR might have a potential neuroprotective effect by promoting cell viability in AD, which might be associated with cell autophagy. Furthermore, MYC, Cdh1, and Acaca might be involved in the progression of AD.
Collapse
|