1
|
Dabas A, Goyal B. Delineating the tryptophan-galactosylamine conjugate mediated structural distortions in Aβ 42 protofibrils. Phys Chem Chem Phys 2025; 27:7336-7355. [PMID: 40123533 DOI: 10.1039/d4cp03330b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Amyloid-β (Aβ) fibrillation into neurotoxic soluble oligomers and mature fibrils is mainly responsible for the etiology of Alzheimer's disease (AD). A recent study revealed 61% disaggregation of the pre-formed Aβ42 fibrils upon incubating with a highly soluble tryptophan-galactosylamine conjugate, WGalNAc. WGalNAc displayed no toxicity and increased the viability of SH-SY5Y cells up to 62.9 ± 2% with an EC50 value of 2.3 μM against Aβ42 pre-formed fibrils. However, the key interactions and disruptive mechanism of WGalNAc against Aβ fibrils remain elusive. Thus, mechanistic insights into the disruptive potential of WGalNAc against Aβ42 protofibrils (PDB: 5OQV) were examined using molecular dynamics (MD) simulations. The molecular docking depicted a favourable binding energy (-6.60 kcal mol-1) and interaction of WGalNAc with the central hydrophobic core (CHC) region of chain A of the 5OQV protofibril. The MD simulations depicted that WGalNAc disrupted the contacts among Ala2, Phe4, Leu34, and Val36 in the hydrophobic core 1 of the 5OQV protofibril responsible for maintaining the stability of the LS-shaped 5OQV protofibril. WGalNAc binds favourably to the 5OQV protofibril (ΔGbinding = -21.76 ± 2.40 kcal mol-1) with a significant contribution from the van der Waals interaction term. Notably, the binding affinity between the neighbouring chains of the 5OQV protofibril was significantly reduced from -134.31 ± 11.12 to -121.88 ± 1.95 kcal mol-1 upon the incorporation of WGalNAc, which is consistent with the ThT kinetic results that revealed disaggregation of the pre-formed Aβ42 fibrils upon incubating with WGalNAc. The in silico ADMET properties of WGalNAc showed its ability as a promising therapeutic candidate due to its blood-brain barrier (BBB) permeability, extended half-life, and non-toxic profile. The MD simulations illuminated the binding interactions of WGalNAc with the 5OQV protofibril and provided mechanistic insights into the WGalNAc-mediated structural distortions in the 5OQV protofibril.
Collapse
Affiliation(s)
- Arushi Dabas
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, 147004, Punjab, India.
| | - Bhupesh Goyal
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, 147004, Punjab, India.
| |
Collapse
|
2
|
Piskorz T, Perez-Chirinos L, Qiao B, Sasselli IR. Tips and Tricks in the Modeling of Supramolecular Peptide Assemblies. ACS OMEGA 2024; 9:31254-31273. [PMID: 39072142 PMCID: PMC11270692 DOI: 10.1021/acsomega.4c02628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/30/2024]
Abstract
Supramolecular peptide assemblies (SPAs) hold promise as materials for nanotechnology and biomedicine. Although their investigation often entails adapting experimental techniques from their protein counterparts, SPAs are fundamentally distinct from proteins, posing unique challenges for their study. Computational methods have emerged as indispensable tools for gaining deeper insights into SPA structures at the molecular level, surpassing the limitations of experimental techniques, and as screening tools to reduce the experimental search space. However, computational studies have grappled with issues stemming from the absence of standardized procedures and relevant crystal structures. Fundamental disparities between SPAs and protein simulations, such as the absence of experimentally validated initial structures and the importance of the simulation size, number of molecules, and concentration, have compounded these challenges. Understanding the roles of various parameters and the capabilities of different models and simulation setups remains an ongoing endeavor. In this review, we aim to provide readers with guidance on the parameters to consider when conducting SPA simulations, elucidating their potential impact on outcomes and validity.
Collapse
Affiliation(s)
| | - Laura Perez-Chirinos
- Center
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014 Donostia-San Sebastián, Spain
| | - Baofu Qiao
- Department
of Natural Sciences, Baruch College, City
University of New York, New York, New York 10010, United States
| | - Ivan R. Sasselli
- Centro
de Física de Materiales (CFM), CSIC-UPV/EHU, Paseo Manuel de Lardizabal 5, 20018 San Sebastián, Spain
| |
Collapse
|
3
|
Chai K, Yang J, Tu Y, Wu J, Fang K, Shi S, Yao T. Molecular Deformation Is a Key Factor in Screening Aggregation Inhibitor for Intrinsically Disordered Protein Tau. ACS CENTRAL SCIENCE 2024; 10:717-728. [PMID: 38559297 PMCID: PMC10979476 DOI: 10.1021/acscentsci.3c01196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 02/03/2024] [Accepted: 02/16/2024] [Indexed: 04/04/2024]
Abstract
Direct inhibitor of tau aggregation has been extensively studied as potential therapeutic agents for Alzheimer's disease. However, the natively unfolded structure of tau complicates the structure-based ligand design, and the relatively large surface areas that mediate tau-tau interactions in aggregation limit the potential for identifying high-affinity ligand binding sites. Herein, a group of isatin-pyrrolidinylpyridine derivative isomers (IPP1-IPP4) were designed and synthesized. They are like different forms of molecular "transformers". These isatin isomers exhibit different inhibitory effects on tau self-aggregation or even possess a depolymerizing effect. Our results revealed for the first time that the direct inhibitor of tau protein aggregation is not only determined by the previously reported conjugated structure, substituent, hydrogen bond donor, etc. but also depends more importantly on the molecular shape. In combination with molecular docking and molecular dynamics simulations, a new inhibition mechanism was proposed: like a "molecular clip", IPP1 could noncovalently bind and fix a tau polypeptide chain at a multipoint to prevent the transition from the "natively unfolded conformation" to the "aggregation competent conformation" before nucleation. At the cellular and animal levels, the effectiveness of the inhibitor of the IPP1 has been confirmed, providing an innovative design strategy as well as a lead compound for Alzheimer's disease drug development.
Collapse
Affiliation(s)
- Keke Chai
- School
of Chemical Science and Engineering, Shanghai Key Laboratory of Chemical
Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Jian Yang
- School
of Medicine, Shanghai University, Shanghai 200444, China
| | - Ying Tu
- School
of Chemical Science and Engineering, Shanghai Key Laboratory of Chemical
Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Junjie Wu
- School
of Chemical Science and Engineering, Shanghai Key Laboratory of Chemical
Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Kang Fang
- School
of Chemical Science and Engineering, Shanghai Key Laboratory of Chemical
Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Shuo Shi
- School
of Chemical Science and Engineering, Shanghai Key Laboratory of Chemical
Assessment and Sustainability, Tongji University, Shanghai 200092, China
| | - Tianming Yao
- School
of Chemical Science and Engineering, Shanghai Key Laboratory of Chemical
Assessment and Sustainability, Tongji University, Shanghai 200092, China
| |
Collapse
|
4
|
Wang T, Ménard-Moyon C, Bianco A. Structural Transformation of Coassembled Fmoc-Protected Aromatic Amino Acids to Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:10532-10544. [PMID: 38367060 DOI: 10.1021/acsami.3c18463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
Materials made of assembled biomolecules such as amino acids have drawn much attention during the past decades. Nevertheless, research on the relationship between the chemical structure of building block molecules, supramolecular interactions, and self-assembled structures is still necessary. Herein, the self-assembly and the coassembly of fluorenylmethoxycarbonyl (Fmoc)-protected aromatic amino acids (tyrosine, tryptophan, and phenylalanine) were studied. The individual self-assembly of Fmoc-Tyr-OH and Fmoc-Phe-OH in water formed nanofibers, while Fmoc-Trp-OH self-assembled into nanoparticles. Moreover, when Fmoc-Tyr-OH or Fmoc-Phe-OH was coassembled with Fmoc-Trp-OH, the nanofibers were transformed into nanoparticles. UV-vis spectroscopy, Fourier transform infrared spectroscopy, and fluorescence spectroscopy were used to investigate the supramolecular interactions leading to the self-assembled architectures. π-π stacking and hydrogen bonding were the main driving forces leading to the self-assembly of Fmoc-Tyr-OH and Fmoc-Phe-OH forming nanofibers. Further, a mechanism involving a two-step coassembly process is proposed based on nucleation and elongation/growth to explain the structural transformation. Fmoc-Trp-OH acted as a fiber inhibitor to alter the molecular interactions in the Fmoc-Tyr-OH or Fmoc-Phe-OH self-assembled structures during the coassembly process, locking the coassembly in the nucleation step and preventing the formation of nanofibers. This structural transformation is useful for extending the application of amino acid self- or coassembled materials in different fields. For example, the amino acids forming nanofibers could be applied for tissue engineering, while they could be exploited as drug nanocarriers when they form nanoparticles.
Collapse
Affiliation(s)
- Tengfei Wang
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000 Strasbourg, France
| | - Cécilia Ménard-Moyon
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000 Strasbourg, France
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR 3572, University of Strasbourg, ISIS, 67000 Strasbourg, France
| |
Collapse
|
5
|
Dolai G, Shill S, Roy S, Mandal B. Atomic Insight on Inhibition of Fibrillization of Dipeptides by Replacement of Phenylalanine with Tryptophan. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023. [PMID: 37339161 DOI: 10.1021/acs.langmuir.3c00823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Tryptophan (Trp) conjugates destabilize amyloid fibrils responsible for amyloidoses. However, the mechanism of such destabilization is obscure. Herein the self-assembly of four synthesized Trp-containing dipeptides Boc-xxx-Trp-OMe (xxx: Val, Leu, Ile, and Phe) has been investigated and compared with the existing report on their Phe congeners. Two among them are the C-terminal tryptophan analogs of Boc-Val-Phe-OMe (VF, Aβ18-19) and Boc-Phe-Phe-OMe (FF, Aβ19-20), part of the central hydrophobic region of amyloid-β (Aβ1-42). While Boc-Val-Trp-OMe (VW), Boc-Leu-Trp-OMe (LW), Boc-Ile-Trp-OMe (IW), and Boc-Phe-Trp-OMe (FW) displayed a spherical morphology in FESEM and AFM images, the corresponding phenylalanine-containing dipeptides displayed various fibrous structures. Single-crystal X-ray diffraction (SC-XRD) indicated that peptides VW and IW exhibited structures containing parallel β-sheet, cross-β-structure, sheet-like layer structure, and helical arrangement in the solid state. Interestingly, peptide FW displayed inverse γ-turn conformation (similar to open-turn structure), antiparallel β-sheet structure, columnar structure, supramolecular nanozipper structure, sheet-like layer arrangement, and helical architecture in the solid state. The open-turn conformation and nanozipper structure formation by FW may be the first example of a dipeptide that forms such structures. The minute but consistent differences in molecular packing at the atomic level between Trp and Phe congeners may be responsible for their remarkably different supramolecular structure generation. This molecular-level structural analysis may be helpful for the de novo design of peptide nanostructures and therapeutics. Similar studies by the Debasish Haldar group are reported, but they investigated the inhibition of fibrillization of dipeptides by tyrosine and interactions are expectedly different.
Collapse
Affiliation(s)
- Gobinda Dolai
- Department of Chemistry, Laboratory of Peptide and Amyloid Research, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sukesh Shill
- Department of Chemistry, Laboratory of Peptide and Amyloid Research, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sayanta Roy
- Department of Chemistry, Laboratory of Peptide and Amyloid Research, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Bhubaneswar Mandal
- Department of Chemistry, Laboratory of Peptide and Amyloid Research, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
6
|
Pal S, Roy R, Paul S. Deciphering the Role of ATP on PHF6 Aggregation. J Phys Chem B 2022; 126:4761-4775. [PMID: 35759245 DOI: 10.1021/acs.jpcb.2c01768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The aggregation of Tau protein, which are involved in Alzheimer's disease, are associated with the self-assembly of the hexapeptide sequence, paired helical filament 6 (PHF6) from repeat 3 of Tau. In order to treat Alzheimer's disease and other such tauopathies, one of the therapeutic strategies is to inhibit aggregation of Tau and its nucleating segments. Therefore, we have studied the effect of adenosine triphosphate (ATP) on the aggregation of PHF6. ATP has, interestingly, demonstrated its ability to inhibit and dissolve protein aggregates. Using classical molecular dynamics simulations, we observed that the hydrophobic core of PHF6 segment displays extended β-sheet conformation, which stabilizes PHF6 aggregates. However, the distribution of ATP around the vicinity of the peptides enables PHF6 to remain discrete and attain random coil conformers. The interpeptide interactions are substituted by PHF6-ATP interactions through hydrogen bonding and hydrophobic interactions (including π-π stacking). Furthermore, the adenosine moiety of ATP contributes more than the triphosphate chain toward PHF6-ATP interaction. Ultimately, this work establishes the inhibitory activity of ATP against Tau aggregation; hence, the therapeutic effect of ATP should be explored further in regard to the effective treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Saikat Pal
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Rituparna Roy
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| |
Collapse
|
7
|
Arad E, Jelinek R, Rapaport H. Amyloid fishing: β-Amyloid adsorption using tailor-made coated titania nanoparticles. Colloids Surf B Biointerfaces 2022; 212:112374. [PMID: 35121429 DOI: 10.1016/j.colsurfb.2022.112374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/19/2022]
Abstract
Amyloidoses are a family of diseases characterized by abnormal protein folding that leads to fibril aggregates, amyloids. Extensive research efforts are devoted to developing inhibitors to amyloid aggregates. Here we set to explore functionalized titania (TiO2) nanoparticles (NPs) as potential amyloid inhibiting agents. TiO2 NPs were coated by a catechol derivative, dihydroxy-phenylalanine propanoic acid (DPA), and further conjugated to the amyloids' specific dye Congo-Red (CR). TiO2-DPA-CR NPs were found to target mature fibrils of β-amyloid (Aβ). Moreover, coated NPs incubated with Aβ proteins suppressed amyloid fibrillation. TiO2-DPA-CR were found to target amyloids in solution and induce their sedimentation upon centrifugation. This work demonstrates the potential utilization of TiO2-DPA NPs for labeling and facilely separating from solution mature amyloid fibrils.
Collapse
Affiliation(s)
- Elad Arad
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Raz Jelinek
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Hanna Rapaport
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
8
|
Paul A, Segal D, Zacco E. Glycans to improve efficacy and solubility of protein aggregation inhibitors. Neural Regen Res 2021; 16:2215-2216. [PMID: 33818499 PMCID: PMC8354138 DOI: 10.4103/1673-5374.310688] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/28/2020] [Accepted: 01/30/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Ashim Paul
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Ramat Aviv, Tel Aviv 6997801, Israel
| | - Daniel Segal
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Ramat Aviv, Tel Aviv 6997801, Israel
| | - Elsa Zacco
- RNA Central Lab, Center for Human Technologies, Istituto Italiano di Tecnologia, 16152 Genova, Italy
| |
Collapse
|
9
|
Li Y, Peng X. Comparison of the force fields on monomeric and fibrillar PHF6 of tau protein. Biophys Chem 2021; 277:106631. [PMID: 34116358 DOI: 10.1016/j.bpc.2021.106631] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/18/2021] [Accepted: 06/01/2021] [Indexed: 11/19/2022]
Abstract
The hexapeptide 306VQIVYK311 (PHF6) plays an important role in the aggregation of Tau protein, which is a hallmark of the Alzheimer's disease (AD). In this article, we systematically compare the effects of eight popular all-atom force fields on the monomeric and fibrillar PHF6 in the molecular dynamics (MD) simulations, which could be helpful in the computer-aided drug design against PHF6. We show that the fibrillar PHF6 prefers β-strand-like structures in all the force fields while the monomer has different structural preferences depending on the force fields. The interactions for stabilizing the fibril are further investigated. In the end, according to the interactions revealed by NMR and the stability of the fibril in the literature, we benchmark the force fields.
Collapse
Affiliation(s)
- Yanchun Li
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Xubiao Peng
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China; Beijing Academy of Quantum Information Sciences, Beijing 100193, China.
| |
Collapse
|
10
|
A systematic review of carbohydrate-based bioactive molecules for Alzheimer's disease. Future Med Chem 2021; 13:1695-1711. [PMID: 34472382 DOI: 10.4155/fmc-2021-0109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The abundance, low cost, high density of functional groups and ease of purification of carbohydrates are among the most important features that make them a prime candidate for designing therapeutics. Several carbohydrate-based molecules, of both natural and synthetic origin, are known for their wide range of therapeutic activities. The incorporation of a carbohydrate moiety not only retains the pharmacological characteristics of a molecule but also improves its activity. Several sugar conjugates have been designed and reported to inhibit acetylcholinesterase, β-amyloid and tau aggregation. This systematic review provides a brief overview of carbohydrate-based bioactive molecules having anti-Alzheimer's activity along with improved therapeutic potential. Most importantly, several reported carbohydrate-based molecules for Alzheimer's disease act on β-amyloid aggregation, tau protein, cholinesterase and oxidative stress, with enhanced pharmacokinetic and mechanistic properties. The prospect of designing carbohydrate-based molecules for Alzheimer's disease will definitely provide potential opportunities to discover novel carbohydrate-based drugs.
Collapse
|
11
|
Li G, Zhou Y, Yang WY, Zhang C, Hong L, Jia L. Inhibitory Effects of Sulfated Polysaccharides from the Sea Cucumber Cucumaria Frondosa against Aβ40 Aggregation and Cytotoxicity. ACS Chem Neurosci 2021; 12:1854-1859. [PMID: 33999600 DOI: 10.1021/acschemneuro.1c00223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Abnormal aggregation and deposition of Aβ is one of the causative agents for Alzheimer's disease. The development of inhibitors for Aβ aggregation has been considered a possible method to prevent and treat Alzheimer's disease. Edible sea cucumbers contain many bioactive molecules, including saponins, phospholipids, peptides, and polysaccharides. Herein, we report that polysaccharides extracted from sea cucumber Cucumaria frondosa could reduce the aggregation and cytotoxicity of Aβ40. By utilizing multiple biochemical and biophysical instruments, we found that the polysaccharides could inhibit the aggregation of Aβ40. A chemical kinetics analysis further suggested that the major inhibitory effects of the polysaccharides were achieved by disassembling mature fibrils, which in turn reduced the cytotoxicity of Aβ. These results suggested that the polysaccharides extracted from sea cucumber could be used as an effective inhibitor for Aβ.
Collapse
Affiliation(s)
- Gao Li
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| | - Yu Zhou
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| | - Wu-Yue Yang
- Yau Mathematical Sciences Center, Tsinghua University, Beijing, Beijing 100084, China
| | - Chen Zhang
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| | - Liu Hong
- School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Lee Jia
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| |
Collapse
|
12
|
Araújo AR, Castro VI, Reis RL, Pires RA. Glucosamine and Its Analogues as Modulators of Amyloid-β Toxicity. ACS Med Chem Lett 2021; 12:548-554. [PMID: 33859794 PMCID: PMC8040036 DOI: 10.1021/acsmedchemlett.0c00350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 03/17/2021] [Indexed: 01/04/2023] Open
Abstract
In Alzheimer's disease (AD), amyloid-β (Aβ) oligomers are considered key mediators of synaptic dysfunction and cognitive impairment. These unstable intermediate Aβ species can interfere with different cellular organelles, leading to neuronal cell death, through the formation of Ca2+-permeable membrane pores, impairment in the levels of acetylcholine neurotransmitters, increased insulin resistance, promotion of pro-inflammatory cascades, among others. Based on a series of evidences that indicate the key role of glycosaminoglycans (GAGs) in amyloid plaque formation, we evaluated the capacity of four monosaccharides, i.e., glucosamine (GlcN), N-acetyl glucosamine (GlcNAc), glucosamine-6-sulfate (GlcN6S), and glucosamine-6-phosphate (GlcN6P), to reduce the Aβ-mediated pathological hallmarks. The tested monosaccharides, in particular, GlcN6S and GlcN6P, were able to interact with Aβ aggregates, reducing neuronal cell death, Aβ-mediated damage to the cellular membrane, acetylcholinesterase activity, insulin resistance, and pro-inflammation levels.
Collapse
Affiliation(s)
- Ana R. Araújo
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials,
Biodegradables and Biomimetics, University
of Minho, Headquarters of the European
Institute of Excellence on Tissue Engineering and Regenerative Medicine,
AvePark, Parque de Ciência e Tecnologia, Zona Industrial da
Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s
− PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Vânia I.
B. Castro
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials,
Biodegradables and Biomimetics, University
of Minho, Headquarters of the European
Institute of Excellence on Tissue Engineering and Regenerative Medicine,
AvePark, Parque de Ciência e Tecnologia, Zona Industrial da
Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s
− PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials,
Biodegradables and Biomimetics, University
of Minho, Headquarters of the European
Institute of Excellence on Tissue Engineering and Regenerative Medicine,
AvePark, Parque de Ciência e Tecnologia, Zona Industrial da
Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s
− PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Ricardo A. Pires
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials,
Biodegradables and Biomimetics, University
of Minho, Headquarters of the European
Institute of Excellence on Tissue Engineering and Regenerative Medicine,
AvePark, Parque de Ciência e Tecnologia, Zona Industrial da
Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s
− PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| |
Collapse
|
13
|
Paul A, Viswanathan GK, Huber A, Arad E, Engel H, Jelinek R, Gazit E, Segal D. Inhibition of tau amyloid formation and disruption of its preformed fibrils by Naphthoquinone-Dopamine hybrid. FEBS J 2021; 288:4267-4290. [PMID: 33523571 DOI: 10.1111/febs.15741] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/02/2021] [Accepted: 01/28/2021] [Indexed: 01/10/2023]
Abstract
Misfolding and aggregation of tau protein, into pathological amyloids, are hallmarks of a group of neurodegenerative diseases collectively termed tauopathies and their modulation may be therapeutically valuable. Herein, we describe the synthesis and characterization of a dopamine-based hybrid molecule, naphthoquinone-dopamine (NQDA). Using thioflavin S assay, CD, transmission electron microscopy, dynamic light scattering, Congo Red birefringence, and large unilamellar vesicle leakage assays, we demonstrated its efficacy in inhibiting the in vitro aggregation of key tau-derived amyloidogenic fragments, PHF6 (VQIVYK) and PHF6* (VQIINK), prime drivers of aggregation of full-length tau in disease pathology. Isothermal titration calorimetry analysis revealed that the interaction between NQDA and PHF6 is spontaneous and has significant binding efficiency driven by both entropic and enthalpic processes. Furthermore, NQDA efficiently disassembled preformed fibrils of PHF6 and PHF6* into nontoxic species. Molecular dynamic simulations supported the in vitro results and provided a plausible mode of binding of NQDA with PHF6 fibril. NQDA was also capable of inhibiting the aggregation of full-length tau protein and disrupting its preformed fibrils in vitro in a dose-dependent manner. In a comparative study, the IC50 value (50% inhibition of fibril formation) of NQDA in inhibiting the aggregation of PHF6 (25 µm) was ~ 17 µm, which is lower than for other bona fide amyloid inhibitors, naphthoquinone-tryptophan, rosmarinic acid, epigallocatechin gallate, ~ 21, ~ 77, or ~ 19 µm, respectively. Comparable superiority of NQDA was observed for inhibition of PHF6*. These findings suggest that NQDA can be a useful scaffold for designing new therapeutics for Alzheimer's disease and other tauopathies.
Collapse
Affiliation(s)
- Ashim Paul
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel
| | - Guru KrishnaKumar Viswanathan
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel
| | - Adi Huber
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel
| | - Elad Arad
- Ilse Katz Institute for Nanoscale Science and Technology & Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Hamutal Engel
- Blavatnik Center for Drug Discovery, Tel Aviv University, Israel
| | - Raz Jelinek
- Ilse Katz Institute for Nanoscale Science and Technology & Department of Chemistry, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel.,Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Israel
| | - Daniel Segal
- Department of Molecular Microbiology and Biotechnology, Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Israel.,Sagol Interdisciplinary School of Neuroscience, Tel Aviv University, Israel
| |
Collapse
|
14
|
Paul A, Frenkel-Pinter M, Escobar Alvarez D, Milordini G, Gazit E, Zacco E, Segal D. Tryptophan-galactosylamine conjugates inhibit and disaggregate amyloid fibrils of Aβ42 and hIAPP peptides while reducing their toxicity. Commun Biol 2020; 3:484. [PMID: 32879439 PMCID: PMC7468108 DOI: 10.1038/s42003-020-01216-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
Self-assembly of proteins into amyloid fibrils is a hallmark of various diseases, including Alzheimer's disease (AD) and Type-2 diabetes Mellitus (T2DM). Aggregation of specific peptides, like Aβ42 in AD and hIAPP in T2DM, causes cellular dysfunction resulting in the respective pathology. While these amyloidogenic proteins lack sequence homology, they all contain aromatic amino acids in their hydrophobic core that play a major role in their self-assembly. Targeting these aromatic residues by small molecules may be an attractive approach for inhibiting amyloid aggregation. Here, various biochemical and biophysical techniques revealed that a panel of tryptophan-galactosylamine conjugates significantly inhibit fibril formation of Aβ42 and hIAPP, and disassemble their pre-formed fibrils in a dose-dependent manner. They are also not toxic to mammalian cells and can reduce the cytotoxicity induced by Aβ42 and hIAPP aggregates. These tryptophan-galactosylamine conjugates can therefore serve as a scaffold for the development of therapeutics towards AD and T2DM.
Collapse
Affiliation(s)
- Ashim Paul
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Moran Frenkel-Pinter
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Daniela Escobar Alvarez
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Giulia Milordini
- The Maurice Wohl Clinical Neuroscience Institute, King's College London, Brixton, London, SE5 9RT, UK
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel
| | - Elsa Zacco
- The Maurice Wohl Clinical Neuroscience Institute, King's College London, Brixton, London, SE5 9RT, UK.
- RNA Central Lab, Center for Human Technologies, Istituto Italiano di Tecnologia, 16152, Genova, Italy.
| | - Daniel Segal
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel.
- Sagol Interdisciplinary School of Neuroscience, Tel Aviv University, Ramat Aviv, Tel Aviv, 6997801, Israel.
| |
Collapse
|
15
|
Li G, Yang W, Li W, Luo Y, Lim Y, Li Y, Paul A, Segal D, Hong L, Li Y. Rational Design of a Cocktail of Inhibitors against Aβ Aggregation. Chemistry 2020; 26:3499-3503. [DOI: 10.1002/chem.201905621] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/21/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Gao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
- Institute of OceanographyMinjiang University 350108 Fuzhou P. R. China
| | - Wu‐Yue Yang
- Zhou Pei-Yuan Center for Applied MathematicsTsinghua University 100084 Beijing P. R. China
| | - Wen‐Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
| | - Yun‐Yi Luo
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
| | - Yeh‐Jun Lim
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
| | - Yang Li
- Institute for Science and TechnologyShandong University of, Traditional Chinese Medicine 250355 Jinan P. R. China
| | - Ashim Paul
- School of Molecular Microbiology & BiotechnologyTel Aviv University 69978 Tel Aviv Israel
| | - Daniel Segal
- School of Molecular Microbiology & BiotechnologyTel Aviv University 69978 Tel Aviv Israel
- Sagol Interdisciplinary School of NeurosciencesTel Aviv University 69978 Tel Aviv Israel
| | - Liu Hong
- Zhou Pei-Yuan Center for Applied MathematicsTsinghua University 100084 Beijing P. R. China
| | - Yan‐Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education)Department of ChemistryTsinghua University 100084 Beijing P. R. China
- Beijing Institute for Brain Disorders 100069 Beijing P. R. China
- Center for Synthetic and Systems BiologyTsinghua University 100084 Beijing P. R. China
| |
Collapse
|