ACS Publications. Most Trusted. Most Cited. Most Read
My Activity
CONTENT TYPES

Figure 1Loading Img

Design of β-Amyloid Aggregation Inhibitors from a Predicted Structural Motif

View Author Information
Department of Chemistry, Stanford University, Stanford, California 94305, United States
Department of Neurology, UCLA, Los Angeles, California 90095, United States
§ Department of Pathology, Stanford University, Stanford, California 94305, United States
Brain Research Institute, UCLA, Los Angeles, California 90095, United States
Molecular Biology Institute, UCLA, Los Angeles, California 90095, United States
*Phone: (650)723-3660. E-mail: [email protected]. Address: Department of Chemistry, Stanford University, 318 West Campus Drive, Stanford, California 94305.
Cite this: J. Med. Chem. 2012, 55, 7, 3002–3010
Publication Date (Web):March 15, 2012
https://doi.org/10.1021/jm201332p
Copyright © 2012 American Chemical Society

    Article Views

    2727

    Altmetric

    -

    Citations

    51
    LEARN ABOUT THESE METRICS
    Other access options
    Supporting Info (1)»

    Abstract

    Abstract Image

    Drug design studies targeting one of the primary toxic agents in Alzheimer’s disease, soluble oligomers of amyloid β-protein (Aβ), have been complicated by the rapid, heterogeneous aggregation of Aβ and the resulting difficulty to structurally characterize the peptide. To address this, we have developed [Nle35, d-Pro37]Aβ42, a substituted peptide inspired from molecular dynamics simulations which forms structures stable enough to be analyzed by NMR. We report herein that [Nle35, d-Pro37]Aβ42 stabilizes the trimer and prevents mature fibril and β-sheet formation. Further, [Nle35, d-Pro37]Aβ42 interacts with WT Aβ42 and reduces aggregation levels and fibril formation in mixtures. Using ligand-based drug design based on [Nle35, d-Pro37]Aβ42, a lead compound was identified with effects on inhibition similar to the peptide. The ability of [Nle35, d-Pro37]Aβ42 and the compound to inhibit the aggregation of Aβ42 provides a novel tool to study the structure of Aβ oligomers. More broadly, our data demonstrate how molecular dynamics simulation can guide experiment for further research into AD.

    Read this article

    To access this article, please review the available access options below.

    Get instant access

    Purchase Access

    Read this article for 48 hours. Check out below using your ACS ID or as a guest.

    Recommended

    Access through Your Institution

    You may have access to this article through your institution.

    Your institution does not have access to this content. You can change your affiliated institution below.

    Supporting Information

    ARTICLE SECTIONS
    Jump To

    Results from experiments conducted with Aβscramble and [Sem35,d-Pro37]Aβ42. LC-MS and 1H NMR of 1, 2, and 3. Rankings and structures of top scoring compounds from the virtual screen. This material is available free of charge via the Internet at http://pubs.acs.org.

    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

    Cited By

    This article is cited by 51 publications.

    1. Gagandeep Kaur, Opinder Kaur Mankoo, Deepti Goyal, Bhupesh Goyal. Unveiling How Hydroxytyrosol Destabilizes α-Syn Oligomers Using Molecular Simulations. The Journal of Physical Chemistry B 2023, 127 (25) , 5620-5632. https://doi.org/10.1021/acs.jpcb.3c02434
    2. Shaoying Xu, Yan Sun, Xiaoyan Dong. Design of Gallic Acid–Glutamine Conjugate and Chemical Implications for Its Potency Against Alzheimer’s Amyloid-β Fibrillogenesis. Bioconjugate Chemistry 2022, 33 (4) , 677-690. https://doi.org/10.1021/acs.bioconjchem.2c00073
    3. Babak Saffari, Mehriar Amininasab. Crocin Inhibits the Fibrillation of Human α-synuclein and Disassembles Mature Fibrils: Experimental Findings and Mechanistic Insights from Molecular Dynamics Simulation. ACS Chemical Neuroscience 2021, 12 (21) , 4037-4057. https://doi.org/10.1021/acschemneuro.1c00379
    4. Wenjuan Wang, Wei Liu, Shaoying Xu, Xiaoyan Dong, Yan Sun. Design of Multifunctional Agent Based on Basified Serum Albumin for Efficient In Vivo β-Amyloid Inhibition and Imaging. ACS Applied Bio Materials 2020, 3 (5) , 3365-3377. https://doi.org/10.1021/acsabm.0c00295
    5. Mantas Ziaunys, Vytautas Smirnovas. Additional Thioflavin-T Binding Mode in Insulin Fibril Inner Core Region. The Journal of Physical Chemistry B 2019, 123 (41) , 8727-8732. https://doi.org/10.1021/acs.jpcb.9b08652
    6. Amandeep Kaur, Simranjeet Singh Narang, Anupamjeet Kaur, Sukhmani Mann, Nitesh Priyadarshi, Bhupesh Goyal, Nitin Kumar Singhal, Deepti Goyal. Multifunctional Mono-Triazole Derivatives Inhibit Aβ42 Aggregation and Cu2+-Mediated Aβ42 Aggregation and Protect Against Aβ42-Induced Cytotoxicity. Chemical Research in Toxicology 2019, 32 (9) , 1824-1839. https://doi.org/10.1021/acs.chemrestox.9b00168
    7. Anjali Jha, Mothukuri Ganesh Kumar, Hosahudya N. Gopi, and Kishore M. Paknikar . Inhibition of β-Amyloid Aggregation through a Designed β-Hairpin Peptide. Langmuir 2018, 34 (4) , 1591-1600. https://doi.org/10.1021/acs.langmuir.7b03617
    8. Deepti Goyal, Suniba Shuaib, Sukhmani Mann, and Bhupesh Goyal . Rationally Designed Peptides and Peptidomimetics as Inhibitors of Amyloid-β (Aβ) Aggregation: Potential Therapeutics of Alzheimer’s Disease. ACS Combinatorial Science 2017, 19 (2) , 55-80. https://doi.org/10.1021/acscombsci.6b00116
    9. Jessica Nasica-Labouze, Phuong H. Nguyen, Fabio Sterpone, Olivia Berthoumieu, Nicolae-Viorel Buchete, Sébastien Coté, Alfonso De Simone, Andrew J. Doig, Peter Faller, Angel Garcia, Alessandro Laio, Mai Suan Li, Simone Melchionna, Normand Mousseau, Yuguang Mu, Anant Paravastu, Samuela Pasquali, David J. Rosenman, Birgit Strodel, Bogdan Tarus, John H. Viles, Tong Zhang, Chunyu Wang, and Philippe Derreumaux . Amyloid β Protein and Alzheimer’s Disease: When Computer Simulations Complement Experimental Studies. Chemical Reviews 2015, 115 (9) , 3518-3563. https://doi.org/10.1021/cr500638n
    10. Justin A. Lemkul and David R. Bevan . The Role of Molecular Simulations in the Development of Inhibitors of Amyloid β-Peptide Aggregation for the Treatment of Alzheimer’s Disease. ACS Chemical Neuroscience 2012, 3 (11) , 845-856. https://doi.org/10.1021/cn300091a
    11. Fumihiko Katagiri, Kazuki Takeyama, Kentaro Hozumi, Yamato Kikkawa, and Motoyoshi Nomizu . Structural Requirement of Fibrogenic Laminin-Derived Peptide A119 (LSNIDYILIKAS) for Amyloid-like Fibril Formation and Cellular Activity. Biochemistry 2012, 51 (41) , 8218-8225. https://doi.org/10.1021/bi300822d
    12. Nathaniel G. N. Milton, Amrutha Chilumuri, Eridan Rocha-Ferreira, Amanda N. Nercessian, and Maria Ashioti . Kisspeptin Prevention of Amyloid-β Peptide Neurotoxicity in Vitro. ACS Chemical Neuroscience 2012, 3 (9) , 706-719. https://doi.org/10.1021/cn300045d
    13. Rajdeep Kaur, Simranjeet Singh Narang, Pritpal Singh, Bhupesh Goyal. Structural and molecular insights into tacrine-benzofuran hybrid induced inhibition of amyloid-β peptide aggregation and BACE1 activity. Journal of Biomolecular Structure and Dynamics 2023, 41 (22) , 13211-13227. https://doi.org/10.1080/07391102.2023.2191722
    14. Apneet Kaur, Bhupesh Goyal. Identification of new pentapeptides as potential inhibitors of amyloid–β42 aggregation using virtual screening and molecular dynamics simulations. Journal of Molecular Graphics and Modelling 2023, 124 , 108558. https://doi.org/10.1016/j.jmgm.2023.108558
    15. Zhongyun Wu, Lianmeng Ye, Nan Yuan, Nuela Manka’a Che Ajuyo, Zhengpan Xiao, Liangwang Liu, Zuqian Chen, Yechun Pei, Yi Min, Dayong Wang. A Molecular Integrative Study on the Inhibitory Effects of WRR and ERW on Amyloid β Peptide (1–42) Polymerization and Cell Toxicity. Polymers 2023, 15 (22) , 4356. https://doi.org/10.3390/polym15224356
    16. Apneet Kaur, Bhupesh Goyal. In silico design and identification of new peptides for mitigating hIAPP aggregation in type 2 diabetes. Journal of Biomolecular Structure and Dynamics 2023, 59 , 1-16. https://doi.org/10.1080/07391102.2023.2254411
    17. Rahul Saini, Govinda R. Navale, Sain Singh, Haobam Kisan Singh, Rahul Chauhan, Sonia Agrawal, Dhiman Sarkar, Manabendra Sarma, Kaushik Ghosh. Inhibition of amyloid β1–42 peptide aggregation by newly designed cyclometallated palladium complexes. International Journal of Biological Macromolecules 2023, 248 , 125847. https://doi.org/10.1016/j.ijbiomac.2023.125847
    18. Vincent A. Voelz, Vijay S. Pande, Gregory R. Bowman. Folding@home: Achievements from over 20 years of citizen science herald the exascale era. Biophysical Journal 2023, 122 (14) , 2852-2863. https://doi.org/10.1016/j.bpj.2023.03.028
    19. Shivani Bagga, Manish Kumar. Current Status of Alzheimer’s Disease and Pathological Mechanisms Investigating the Therapeutic Molecular Targets. Current Molecular Medicine 2023, 23 (6) , 492-508. https://doi.org/10.2174/1566524022666220404112843
    20. Sain Singh, Govinda R. Navale, Sonia Agrawal, Haobam Kisan Singh, Labhini Singla, Dhiman Sarkar, Manabendra Sarma, Anghuman Roy Choudhury, Kaushik Ghosh. Design and synthesis of piano-stool ruthenium(II) complexes and their studies on the inhibition of amyloid β (1–42) peptide aggregation. International Journal of Biological Macromolecules 2023, 239 , 124197. https://doi.org/10.1016/j.ijbiomac.2023.124197
    21. Opinder Kaur Mankoo, Anupamjeet Kaur, Deepti Goyal, Bhupesh Goyal. Unravelling the destabilization potential of ellagic acid on α-synuclein fibrils using molecular dynamics simulations. Physical Chemistry Chemical Physics 2023, 25 (11) , 8128-8143. https://doi.org/10.1039/D2CP06006J
    22. John T. Lawler, Christopher P. Harrilal, Andrew F. DeBlase, Edwin L. Sibert, Scott A. McLuckey, Timothy S. Zwier. Single-conformation spectroscopy of cold, protonated D PG-containing peptides: switching β-turn types and formation of a sequential type II/II′ double β-turn. Physical Chemistry Chemical Physics 2022, 24 (4) , 2095-2109. https://doi.org/10.1039/D1CP04852J
    23. Rajdeep Kaur, Rajneet Kaur Saini, Pritpal Singh, Bhupesh Goyal. Unveiling the inhibitory mechanism of peptidomimetic inhibitor against Aβ42 aggregation and protofibril disaggregation by molecular dynamics. Journal of Molecular Liquids 2021, 335 , 116474. https://doi.org/10.1016/j.molliq.2021.116474
    24. Xicui Sun, Songwei Duan, Anna Cao, Bryan Villagomez, Runxuan Lin, Hongxia Chen, Liya Pi, Bin Ren, Rong Chen, Minjie Chen, Zhekang Ying, Shenyun Fang, Qi Cao. RRY Inhibits Amyloid-β1–42 Peptide Aggregation and Neurotoxicity. Journal of Alzheimer's Disease Reports 2021, 5 (1) , 479-495. https://doi.org/10.3233/ADR-210012
    25. Raúl Horacio Camarillo-López, Maricarmen Hernández Rodríguez, Mónica Adriana Torres-Ramos, Ivonne Maciel Arciniega-Martínez, Iohanan Daniel García-Marín, José Correa Basurto, Juan Vicente Méndez Méndez, Martha Cecilia Rosales-Hernández. Tert-butyl-(4-hydroxy-3-((3-(2-methylpiperidin-yl)propyl)carbamoyl)phenyl)carbamate Has Moderated Protective Activity in Astrocytes Stimulated with Amyloid Beta 1-42 and in a Scopolamine Model. Molecules 2020, 25 (21) , 5009. https://doi.org/10.3390/molecules25215009
    26. Shaochun Shi, Huibin Wang, Jian Wang, Yvxi Wang, Xiaobian Xue, Zilin Hou, Guo-Dong Yao, Xiao-Xiao Huang, Hongwei Zhao, Qingbo Liu, Shao-Jiang Song. Semi-synthesis and biological evaluation of flavone hybrids as multifunctional agents for the potential treatment of Alzheimer's disease. Bioorganic Chemistry 2020, 100 , 103917. https://doi.org/10.1016/j.bioorg.2020.103917
    27. Varnavas D. Mouchlis, Georgia Melagraki, Lefteris C. Zacharia, Antreas Afantitis. Computer-Aided Drug Design of β-Secretase, γ-Secretase and Anti-Tau Inhibitors for the Discovery of Novel Alzheimer’s Therapeutics. International Journal of Molecular Sciences 2020, 21 (3) , 703. https://doi.org/10.3390/ijms21030703
    28. Asmaa Awad, Hedia Fgaier, Ibrahim Mustafa, Ali Elkamel, Said Elnashaie. Pharmacokinetic/Pharmacodynamic modeling and simulation of the effect of medications on β-amyloid aggregates and cholinergic neurocycle. Computers & Chemical Engineering 2019, 126 , 231-240. https://doi.org/10.1016/j.compchemeng.2019.04.017
    29. Anjali Jha, Vandana Ghormade, Henry Kolge, Kishore M. Paknikar. Dual effect of chitosan-based nanoparticles on the inhibition of β-amyloid peptide aggregation and disintegration of the preformed fibrils. Journal of Materials Chemistry B 2019, 7 (21) , 3362-3373. https://doi.org/10.1039/C9TB00162J
    30. Simranjeet Singh Narang, Deepti Goyal, Bhupesh Goyal. Inhibition of Alzheimer’s amyloid-β 42 peptide aggregation by a bi-functional bis-tryptoline triazole: key insights from molecular dynamics simulations. Journal of Biomolecular Structure and Dynamics 2019, 13 , 1-14. https://doi.org/10.1080/07391102.2019.1614093
    31. Dipanwita Roy, Dipita Bhattacharyya, Anirban Bhunia. Do Catechins (ECG and EGCG) Bind to the Same Site as Thioflavin T (ThT) in Amyloid Fibril? Answer From Saturation Transfer Difference NMR. Natural Product Communications 2019, 14 (5) , 1934578X1984979. https://doi.org/10.1177/1934578X19849791
    32. Qingbo Liu, Jie Wang, Bin Lin, Zhuo-Yang Cheng, Ming Bai, Shaochun Shi, Xiao-Xiao Huang, Shao-Jiang Song. Phenylpropanoids and lignans from Prunus tomentosa seeds as efficient β-amyloid (Aβ) aggregation inhibitors. Bioorganic Chemistry 2019, 84 , 269-275. https://doi.org/10.1016/j.bioorg.2018.11.041
    33. Le Zhou, Jie Wang, Rui Guo, Bin Lin, Xiao-Bo Wang, Xiao-Xiao Huang, Shao-Jiang Song. Discovery of dihydrobenzofuran neolignans from Rubus ideaus L. with enantioselective anti-Aβ1–42 aggregation activity. Bioorganic Chemistry 2018, 80 , 64-69. https://doi.org/10.1016/j.bioorg.2018.05.016
    34. Suniba Shuaib, Bhupesh Goyal. Scrutiny of the mechanism of small molecule inhibitor preventing conformational transition of amyloid-β 42 monomer: insights from molecular dynamics simulations. Journal of Biomolecular Structure and Dynamics 2018, 36 (3) , 663-678. https://doi.org/10.1080/07391102.2017.1291363
    35. Maricarmen Hernández Rodríguez, Leticia Guadalupe Fragoso Morales, José Correa Basurto, Martha Cecilia Rosales Hernández. Molecular Docking and Molecular Dynamics Simulation to Evaluate Compounds That Avoid the Amyloid Beta 1-42 Aggregation. 2018, 229-248. https://doi.org/10.1007/978-1-4939-7404-7_9
    36. Marek Bajda, Sławomir Filipek. Computational approach for the assessment of inhibitory potency against beta-amyloid aggregation. Bioorganic & Medicinal Chemistry Letters 2017, 27 (2) , 212-216. https://doi.org/10.1016/j.bmcl.2016.11.072
    37. Zhidong Liu, Aihua Zhang, Hui Sun, Ying Han, Ling Kong, Xijun Wang. Two decades of new drug discovery and development for Alzheimer's disease. RSC Advances 2017, 7 (10) , 6046-6058. https://doi.org/10.1039/C6RA26737H
    38. Najmeh Mahmoodabadi, Davood Ajloo. QSAR, docking, and Molecular dynamic studies on the polyphenolic as inhibitors of β-amyloid aggregation. Medicinal Chemistry Research 2016, 25 (10) , 2104-2118. https://doi.org/10.1007/s00044-016-1620-0
    39. Hiroyuki Kawashima, Mei Katayama, Ryota Yoshida, Kenichi Akaji, Akiko Asano, Mitsunobu Doi. A dimer model of human calcitonin13-32 forms an α-helical structure and robustly aggregates in 50% aqueous 2,2,2-trifluoroethanol solution. Journal of Peptide Science 2016, 22 (7) , 480-484. https://doi.org/10.1002/psc.2891
    40. Maria Giovanna Di Carlo, Velia Minicozzi, Vito Foderà, Valeria Militello, Valeria Vetri, Silvia Morante, Maurizio Leone. Thioflavin T templates amyloid β(1–40) conformation and aggregation pathway. Biophysical Chemistry 2015, 206 , 1-11. https://doi.org/10.1016/j.bpc.2015.06.006
    41. Maricarmen Hernández-Rodríguez, José Correa-Basurto, María Inés Nicolás-Vázquez, René Miranda-Ruvalcaba, Claudia Guadalupe Benítez-Cardoza, Aldo Arturo Reséndiz-Albor, Juan Vicente Méndez-Méndez, Martha C. Rosales-Hernández, . Virtual and In Vitro Screens Reveal a Potential Pharmacophore that Avoids the Fibrillization of Aβ1–42. PLOS ONE 2015, 10 (7) , e0130263. https://doi.org/10.1371/journal.pone.0130263
    42. Antonio Francioso, Pasqualina Punzi, Alberto Boffi, Clorinda Lori, Sara Martire, Cesare Giordano, Maria D’Erme, Luciana Mosca. β-Sheet interfering molecules acting against β-amyloid aggregation and fibrillogenesis. Bioorganic & Medicinal Chemistry 2015, 23 (8) , 1671-1683. https://doi.org/10.1016/j.bmc.2015.02.041
    43. Andrew J Doig, Philippe Derreumaux. Inhibition of protein aggregation and amyloid formation by small molecules. Current Opinion in Structural Biology 2015, 30 , 50-56. https://doi.org/10.1016/j.sbi.2014.12.004
    44. Zhiren Wang, Yali Wang, Wenrui Li, Zhihong Liu, Zonghua Luo, Yang Sun, Ruibo Wu, Ling Huang, Xingshu Li. Computer-assisted designed “selenoxy–chinolin”: a new catalytic mechanism of the GPx-like cycle and inhibition of metal-free and metal-associated Aβ aggregation. Dalton Transactions 2015, 44 (48) , 20913-20925. https://doi.org/10.1039/C5DT02130H
    45. Silvia Merlo, Erika Sironi, Laura Colombo, Francisco Cardona, Alessandra M. Martorana, Mario Salmona, Barbara La Ferla, Cristina Airoldi. Cis ‐Glyco‐Fused Benzopyran Derivatives as Hit Compounds for the Development of Therapeutic and Diagnostic Tools against Neurodegenerative Diseases. ChemPlusChem 2014, 79 (6) , 835-843. https://doi.org/10.1002/cplu.201400035
    46. Velia Minicozzi, Roberta Chiaraluce, Valerio Consalvi, Cesare Giordano, Claudia Narcisi, Pasqualina Punzi, Giancarlo C. Rossi, Silvia Morante. Computational and Experimental Studies on β-Sheet Breakers Targeting Aβ1–40 Fibrils. Journal of Biological Chemistry 2014, 289 (16) , 11242-11252. https://doi.org/10.1074/jbc.M113.537472
    47. Fei Mao, Jun Yan, Jianheng Li, Xian Jia, Hui Miao, Yang Sun, Ling Huang, Xingshu Li. New multi-target-directed small molecules against Alzheimer's disease: a combination of resveratrol and clioquinol. Organic & Biomolecular Chemistry 2014, 12 (31) , 5936. https://doi.org/10.1039/C4OB00998C
    48. Shuwen Zeng, Dominique Baillargeat, Ho-Pui Ho, Ken-Tye Yong. Nanomaterials enhanced surface plasmon resonance for biological and chemical sensing applications. Chemical Society Reviews 2014, 43 (10) , 3426. https://doi.org/10.1039/c3cs60479a
    49. Craig B. Stevens, James M. Hanna, Robin K. Lammi. Synthesis of tetrahydroxybiphenyls and tetrahydroxyterphenyls and their evaluation as amyloid-β aggregation inhibitors. Bioorganic & Medicinal Chemistry Letters 2013, 23 (6) , 1703-1706. https://doi.org/10.1016/j.bmcl.2013.01.076
    50. Ashok D. Pehere, Christopher J. Sumby, Andrew D. Abell. New cylindrical peptide assemblies defined by extended parallel β-sheets. Org. Biomol. Chem. 2013, 11 (3) , 425-429. https://doi.org/10.1039/C2OB26637G
    51. Jean-Christophe M. Monbaliu, Alan R. Katritzky. Recent trends in Cys- and Ser/Thr-based synthetic strategies for the elaboration of peptide constructs. Chemical Communications 2012, 48 (95) , 11601. https://doi.org/10.1039/c2cc34434c

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    Pair your accounts.

    Export articles to Mendeley

    Get article recommendations from ACS based on references in your Mendeley library.

    You’ve supercharged your research process with ACS and Mendeley!

    STEP 1:
    Click to create an ACS ID

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

    MENDELEY PAIRING EXPIRED
    Your Mendeley pairing has expired. Please reconnect