1
|
Gao Z, Qiu R, Dave DR, Chandravanshi P, Soares GP, Smith CS, Ortega JA, Palmer LC, Álvarez Z, Stupp SI. Supramolecular Copolymerization of Glycopeptide Amphiphiles and Amyloid Peptides Improves Neuron Survival. J Am Chem Soc 2025; 147:17710-17724. [PMID: 40365999 DOI: 10.1021/jacs.5c00105] [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: 05/15/2025]
Abstract
Neurodegenerative diseases such as Alzheimer's disease and amyotrophic lateral sclerosis are characterized by progressive neuronal loss and the accumulation of misfolded proteins including amyloid proteins. Current therapeutic options include the use of antibodies for these proteins, but novel chemical strategies need to be developed. The disaccharide trehalose has been widely reported to prevent misfolding and aggregation of proteins, and we therefore investigated the conjugation of this moiety to biocompatible peptide amphiphiles (TPAs) known to undergo supramolecular polymerization. Using X-ray scattering, circular dichroism, and infrared spectroscopy, we found that trehalose conjugation destabilized the internal β-sheet structures within the TPA supramolecular polymers as evidenced by a lower thermal transition. Thioflavin T fluorescence showed that these metastable TPA nanofibers suppressed A42 aggregation. Interestingly, we found that the suppression involved supramolecular copolymerization of TPA polymers with Aβ42, which effectively trapped the peptides within the filamentous structures. In vitro assays with human induced pluripotent stem cell-derived neurons demonstrated that these TPAs significantly improved neuron survival compared to other conditions. Our study highlights the potential of properly tuned supramolecular polymerizations of monomers to safely remove amyloidogenic proteins in neurodegeneration.
Collapse
Affiliation(s)
- Zijun Gao
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Ruomeng Qiu
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Dhwanit R Dave
- Center for Regenerative Nanomedicine Northwestern University 303 E. Superior Street, Chicago, Illinois 60611, United States
| | - Palash Chandravanshi
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid 28029, Spain
| | - Gisele P Soares
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona L'Hospitalet de Llobregat, Barcelona 08907, Spain
- Neurodevelopmental Disorders Group, NeuroBell, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Cara S Smith
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Regenerative Nanomedicine Northwestern University 303 E. Superior Street, Chicago, Illinois 60611, United States
| | - J Alberto Ortega
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona L'Hospitalet de Llobregat, Barcelona 08907, Spain
- Neurodevelopmental Disorders Group, NeuroBell, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Liam C Palmer
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Center for Regenerative Nanomedicine Northwestern University 303 E. Superior Street, Chicago, Illinois 60611, United States
| | - Zaida Álvarez
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
- Center for Regenerative Nanomedicine Northwestern University 303 E. Superior Street, Chicago, Illinois 60611, United States
- Department of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid 28029, Spain
| | - Samuel I Stupp
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona 08028, Spain
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Center for Regenerative Nanomedicine Northwestern University 303 E. Superior Street, Chicago, Illinois 60611, United States
- Department of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
2
|
Schützmann MP, Hoyer W. Off-pathway oligomers of α-synuclein and Aβ inhibit secondary nucleation of α-synuclein amyloid fibrils. J Mol Biol 2025; 437:169048. [PMID: 40015369 DOI: 10.1016/j.jmb.2025.169048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
α-Synuclein (αSyn) is a key culprit in the pathogenesis of synucleinopathies such as Parkinson's Disease (PD), in which it forms not only insoluble aggregates called amyloid fibrils but also smaller, likely more detrimental species termed oligomers. This property is shared with other amyloidogenic proteins such as the Alzheimer's Disease-associated amyloid-β (Aβ). We previously found an intriguing interplay between off-pathway Aβ oligomers and Aβ fibrils, in which the oligomers interfere with fibril formation via inhibition of secondary nucleation by blocking secondary nucleation sites on the fibril surface. Here, using ThT aggregation kinetics and atomic force microscopy (AFM), we tested if the same interplay applies to αSyn fibrils. Both homotypic (i.e. αSyn) and heterotypic (i.e. Aβ) off-pathway oligomers inhibited αSyn aggregation in kinetic assays of secondary nucleation. Initially soluble, kinetically trapped Aβ oligomers co-precipitated with αSyn(1-108) fibrils. The resulting co-assemblies were imaged as clusters of curvilinear oligomers by AFM. The results indicate that off-pathway oligomers have a general tendency to bind amyloid fibril surfaces, also in the absence of sequence homology between fibril and oligomer. The interplay between off-pathway oligomers and amyloid fibrils adds another level of complexity to the homo- and hetero-assembly processes of amyloidogenic proteins.
Collapse
Affiliation(s)
- Marie P Schützmann
- Institut für Physikalische Biologie, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, 40204 Germany
| | - Wolfgang Hoyer
- Institut für Physikalische Biologie, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, 40204 Germany; Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, Jülich, 52428 Germany.
| |
Collapse
|
3
|
Ralhan J, Hooda G, Nath D, Pal A. Role of Symmetry to Dictate Pathway-Driven Self-assembly in Photoresponsive Peptide Amphiphiles. Chemistry 2025:e202501288. [PMID: 40350384 DOI: 10.1002/chem.202501288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Stimuli-responsive peptides offer interesting structural space in the realm of emergent pathway-driven supramolecular polymerization toward designing functional nanomaterials with precise structure-function characteristics. However, the role of molecular symmetry in such peptides to govern the self-assembly pathway dynamics remains less explored. We design amyloid-inspired, azobenzene-functionalized symmetric (P1) and asymmetric (P2) peptide amphiphiles in the E form of the azobenzene motif to systematically investigate the interplay of molecular symmetry, solvent composition, and temperature to guide supramolecular organization. Spectroscopic and microscopic analyses reveal distinct aggregation pathways, with E-P1 exhibiting rapid nucleation-elongation toward robust and stable nanofibers, while E-P2 assembles into a kinetic state (E-P2(KS)) that subsequently transforms into thermodynamically favored nanoribbons, E-P2(TS), via a "good and bad solvent" modulation strategy. Further, E-P1 remains structurally resilient due to stronger intermolecular interactions. Interestingly, E-P2 demonstrates reversible E→Z photoisomerization in both kinetic and thermodynamic states, facilitating supramolecular disassembly while retaining the self-organization memory. Notably, the activation energy barriers for Z→E isomerization in P2 differ between its kinetic and thermodynamic states, underscoring the role of pathway complexity and molecular symmetry in peptide assembly. These insights advance the understanding of symmetry-regulated supramolecular assembly to modulate the emergent functional properties for potential applications in nanotechnology and biomaterials.
Collapse
Affiliation(s)
- Jahanvi Ralhan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Mohali, Punjab, 140306, India
| | - Gunjan Hooda
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Mohali, Punjab, 140306, India
| | - Debasish Nath
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Mohali, Punjab, 140306, India
| | - Asish Pal
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Mohali, Punjab, 140306, India
| |
Collapse
|
4
|
Wei J, Meisl G, Dear AJ, Michaels TCT, Knowles TPJ. Kinetics of Amyloid Oligomer Formation. Annu Rev Biophys 2025; 54:185-207. [PMID: 39929552 DOI: 10.1146/annurev-biophys-080124-122953] [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] [Indexed: 05/07/2025]
Abstract
Low-molecular-weight oligomers formed from amyloidogenic peptides and proteins have been identified as key cytotoxins across a range of neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. Developing therapeutic strategies that target oligomers is therefore emerging as a promising approach for combating protein misfolding diseases. As such, there is a great need to understand the fundamental properties, dynamics, and mechanisms associated with oligomer formation. In this review, we discuss how chemical kinetics provides a powerful tool for studying these systems. We review the chemical kinetics approach to determining the underlying molecular pathways of protein aggregation and discuss its applications to oligomer formation and dynamics. We discuss how this approach can reveal detailed mechanisms of primary and secondary oligomer formation, including the role of interfaces in these processes. We further use this framework to describe the processes of oligomer conversion and dissociation, and highlight the distinction between on-pathway and off-pathway oligomers. Furthermore, we showcase on the basis of experimental data the diversity of pathways leading to oligomer formation in various in vitro and in silico systems. Finally, using the lens of the chemical kinetics framework, we look at the current oligomer inhibitor strategies both in vitro and in vivo.
Collapse
Affiliation(s)
- Jiapeng Wei
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom; , ,
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom; , ,
| | - Alexander J Dear
- Department of Biology, Institute of Biochemistry, ETH Zurich, Zurich, Switzerland; ,
- Bringing Materials to Life Initiative, ETH Zurich, Zurich, Switzerland
| | - Thomas C T Michaels
- Department of Biology, Institute of Biochemistry, ETH Zurich, Zurich, Switzerland; ,
- Bringing Materials to Life Initiative, ETH Zurich, Zurich, Switzerland
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom; , ,
| |
Collapse
|
5
|
Kimura S, Adachi K, Ishii Y, Komiyama T, Saito T, Nakayama N, Yokoya M, Takaya H, Yagai S, Kawai S, Uchihashi T, Yamanaka M. Molecular-level insights into the supramolecular gelation mechanism of urea derivative. Nat Commun 2025; 16:3758. [PMID: 40263273 PMCID: PMC12015314 DOI: 10.1038/s41467-025-59032-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
Despite being a promising soft material embodied by molecular self-assembly, the formation mechanism of supramolecular gels remains challenging to fully understand. Here we provide molecular to nanoscopic insights into the formation mechanism of gel-forming fibers from a urea derivative. High-speed atomic force microscopy of the urea derivative revealed the presence of a lag phase prior to the formation of supramolecular fibers, suggesting a nucleation process. The fiber growth kinetics differ at both termini of the fiber, indicating a directional hydrogen-bonding motif by the urea units, which is supported by single-crystal X-ray crystallography of a reference compound. Moreover, we observed an intermittent growth pattern of the fibers with repeated elongation and pause phases. This unique behavior can be simulated by a theoretical block-stacking model. A statistical analysis of the concentration-dependent lag time on macroscopic observation of the gelation suggests the presence of a tetrameric or octameric nucleus of the urea molecules.
Collapse
Affiliation(s)
- Shinya Kimura
- Meiji Pharmaceutical University, Kiyose, Tokyo, Japan.
| | - Kurea Adachi
- Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Yoshiki Ishii
- Department of Physics and Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Tomoki Komiyama
- Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
- Department of Chemistry, Faculty of Science, Shizuoka University, Shizuoka, Japan
| | - Takuho Saito
- Division of Advanced Science and Engineering, Graduate School of Science and Engineering, Chiba University, Chiba, Japan
| | | | | | - Hikaru Takaya
- Department of Life Science, Faculty of Life & Environmental Sciences, Teikyo University of Science, Adachi-ku, Tokyo, Japan
- Division of Advanced Molecular Science, Institute for Molecular Science, National Institute of Natural Science, Okazaki, Aichi, Japan
| | - Shiki Yagai
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, Chiba, Japan.
- Institute for Advanced Academic Research (IAAR), Chiba University, Chiba, Japan.
| | - Shinnosuke Kawai
- Department of Chemistry, Faculty of Science, Shizuoka University, Shizuoka, Japan.
| | - Takayuki Uchihashi
- Department of Physics and Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan.
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, Japan.
| | | |
Collapse
|
6
|
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
|
7
|
Pálmadóttir T, Getachew J, Ortigosa-Pascual L, Axell E, Wei J, Olsson U, Knowles TPJ, Linse S. On the reversibility of amyloid fibril formation. BIOPHYSICS REVIEWS 2025; 6:011303. [PMID: 39973975 PMCID: PMC11836874 DOI: 10.1063/5.0236947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 12/31/2024] [Indexed: 02/21/2025]
Abstract
Amyloids are elongated supramolecular protein self-assemblies. Their formation is a non-covalent assembly process and as such is fully reversible. Amyloid formation is associated with several neurodegenerative diseases, and the reversibility is key to maintaining the healthy state. Reversibility is also key to the performance of fibril-based biomaterials and functional amyloids. The reversibility can be observed by a range of spectroscopic, calorimetric, or surface-based techniques using as a starting state either a supersaturated monomer solution or diluted fibrils. Amyloid formation has the characteristics of a phase transition, and we provide some basic formalism for the reversibility and the derivation of the solubility/critical concentration. We also discuss conditions under which the dissociation of amyloids may be so slow that the process can be viewed as practically irreversible, for example, because it is slow relative to the experimental time frame or because the system at hand contains a source for constant monomer addition.
Collapse
Affiliation(s)
| | - Josef Getachew
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | | | - Emil Axell
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Jiapeng Wei
- Yusuf Hamied Chemistry Department, University of Cambridge, Cambridge, United Kingdom
| | - Ulf Olsson
- Physical Chemistry, Lund University, Lund, Sweden
| | - Tuomas P. J. Knowles
- Yusuf Hamied Chemistry Department, University of Cambridge, Cambridge, United Kingdom
| | - Sara Linse
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| |
Collapse
|
8
|
Maji M, Khajanchi S. Mathematical models on Alzheimer's disease and its treatment: A review. Phys Life Rev 2025; 52:207-244. [PMID: 39813887 DOI: 10.1016/j.plrev.2025.01.004] [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: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/18/2025]
Abstract
Alzheimer's disease is a gradually advancing neurodegenerative disease. According to the report by "World Health Organization (WHO)", there are over 55 million individuals currently living with Alzheimer's disease and other dementia globally, and the number of sufferers is increasing every day. In absence of effective cures and preventive measures, this number is predicted to triple by 2050. The disease's origin is still unclear, and also no such treatment is available for eradicating the disease. Based on the crucial factors that are connected to the disease's progression, the authors developed several types of mathematical models. We review such mathematical models that are utilized to better understand the pathophysiology of Alzheimer's disease. Section-wise, we categorize the mathematical models in terms of different components that might be responsible for Alzheimer's disease. We explain the mathematical models with their descriptions and respective conclusions. In addition to mathematical models, we concentrate on biological aspects of the disease and possible therapeutic targets. We explore the disease's biological basis primarily to understand how proteins, glial cells, cytokines, genes, calcium signaling and oxidative stress contribute to the disease. We go through several treatment targets that might stop the progression of the disease or at least slow it down. We present a table that summarizes the mathematical models in terms of their formalisms, highlighting key components and important remarks.
Collapse
Affiliation(s)
- Mitali Maji
- Department of Mathematics, Presidency University, 86/1 College Street, Kolkata 700073, India
| | - Subhas Khajanchi
- Department of Mathematics, Presidency University, 86/1 College Street, Kolkata 700073, India.
| |
Collapse
|
9
|
Higuchi A, Som A, Wakabayashi R, Goto M, Kamiya N, Besenius P. The Impact of Single Amino Acid Insertion on the Supramolecular Assembly Pathway of Aromatic Peptide Amphiphiles. Chemistry 2025; 31:e202404233. [PMID: 39688070 DOI: 10.1002/chem.202404233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/18/2024]
Abstract
Understanding the mechanism of self-assembly driven by non-covalent interactions is crucial for designing supramolecular materials with desired properties. Here we investigate the self-assembly of aromatic peptide amphiphiles, Fmoc-L2QG and Fmoc-L3QG using a combination of spectroscopic, transmission electron and superresolution optical microscopy techniques. Our results show that Fmoc-L2QG leads to concentration-dependent assembly, forming fibrous assemblies at low concentrations and supramolecular droplets via liquid-liquid phase separation (LLPS) at higher concentrations. Mechanical activation using for example ultrasonication triggered the transition from metastable droplets to fibre morphologies of Fmoc-L2QG. In contrast, Fmoc-L3QG followed both on-pathway and off-pathway routes, resulting in the formation of fibrous morphologies regardless of concentration. Seeding experiments revealed that homo-seeds of the same peptide sequence accelerated the on-pathway process, while hetero-seeds of a mismatched peptide sequences accelerated the off-pathway process, highlighting the competing nature of the complex assembly profile. These findings demonstrate the significant impact of single amino acid insertion on the supramolecular assembly process of oligopeptide monomers, and highlight the potential for controlling the structure and dynamics of peptide materials. Pathway engineering of oligopeptide building blocks and multidomain supramolecular monomers will open new avenues in tailor-made and customizable supramolecular biomaterials.
Collapse
Affiliation(s)
- Ayato Higuchi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Arka Som
- Department of Chemistry, Johannes Gutenberg-University Mainz, Duesbergweg 10- 14, D-55128, Mainz, Germany
| | - Rie Wakabayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
- Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
- Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan
| | - Pol Besenius
- Department of Chemistry, Johannes Gutenberg-University Mainz, Duesbergweg 10- 14, D-55128, Mainz, Germany
| |
Collapse
|
10
|
Aubrey LD, Radford SE. How is the Amyloid Fold Built? Polymorphism and the Microscopic Mechanisms of Fibril Assembly. J Mol Biol 2025:169008. [PMID: 39954780 DOI: 10.1016/j.jmb.2025.169008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
For a given protein sequence, many, up to sometimes hundreds of different amyloid fibril folds, can be formed in vitro. Yet, fibrils extracted from, or found in, human tissue, usually at the end of a long disease process, are often structurally homogeneous. Through monitoring of amyloid assembly reactions in vitro, the scientific community has gained a detailed understanding of the kinetic mechanisms of fibril assembly and the rates at which the different processes involved occur. However, how this kinetic information relates to the structural changes as a protein transforms from its initial, native structure to the canonical cross-β structure of amyloid remain obscure. While cryoEM has yielded a plethora of high-resolution information that portray a vast variety of fibril structures, there remains little knowledge of how and why each particular structure resulted. Recent work has demonstrated that fibril structures can change over an assembly time course, despite the different fibril types having similar thermodynamic stability. This points to kinetic control of the fibrils formed, with structures that initiate or elongate faster becoming the dominant products of assembly. Annotating kinetic assembly mechanisms alongside structural analysis of the fibrils formed is required to truly understand the molecular mechanisms of amyloid formation. However, this is a complicated task. In this review, we discuss how embracing this challenge could open new frontiers in amyloid research and new opportunities for therapy.
Collapse
Affiliation(s)
- Liam D Aubrey
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Science, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Science, University of Leeds, Leeds LS2 9JT, United Kingdom.
| |
Collapse
|
11
|
Mavlankar NA, Awasthi AK, Pradhan MK, Chandran Y, Balakrishnan V, Srivastava A, Pal A. Attenuation of Pathway Complexity in Arginine-Rich Peptide with Polydopamine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409176. [PMID: 39718238 DOI: 10.1002/smll.202409176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/18/2024] [Indexed: 12/25/2024]
Abstract
Dynamic peptide networks represent an attractive structural space of supramolecular polymers in the realm of emergent complexity. Point mutations in the peptide sequence exert profound effects over the landscapes of self-assembly with an intricate interplay among the structure-function relationships. Herein, the pathway complexity of an arginine-rich peptide is studied, FmocVFFARR derived by the mutation of minimalist amyloid-inspired peptide amphiphile FmocVFFAKK, thereby focusing on its pathway-dependent self-assembly behavior. Interestingly, an interplay of competing primary and secondary nucleation in this minimalist model presumably due to the sticky interactions of the di-arginine motifs is encountered. This furnishes transient nanosheets from on-pathway metastable nanoparticles upon pH trigger, eventually leading to nanofibers. Moreover, external cues, e.g., pH, and temperature convert the nanofibers in off-pathway nanoparticles. For the first time, polydopamine-based surface engineering strategy to mask the arginines is demonstrated to render permanent arrest of the dynamic, transient peptide nanostructures. Finally, such polydopamine layer over the peptide nanostructures furnishes resilience against environmental stress, while also imparting mechanical robustness to the composites. The dynamic peptide nanostructures exhibited adaptive systems capable of processing chemical information while the surface coated nanostructures open wide avenues for designing stress-resilient biomaterials.
Collapse
Affiliation(s)
- Nimisha A Mavlankar
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Sector 81, Mohali, Punjab, 140306, India
| | - Anand K Awasthi
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Sector 81, Mohali, Punjab, 140306, India
| | - Manas K Pradhan
- Department of Chemistry, IISER Bhopal, Bhopal, Madhya Pradesh, 462066, India
| | - Yadu Chandran
- School of Mechanical and Materials Engineering, Indian Institute of Technology (IIT Mandi), Kamand, Himachal Pradesh, 175005, India
| | - Viswanath Balakrishnan
- School of Mechanical and Materials Engineering, Indian Institute of Technology (IIT Mandi), Kamand, Himachal Pradesh, 175005, India
| | - Aasheesh Srivastava
- Department of Chemistry, IISER Bhopal, Bhopal, Madhya Pradesh, 462066, India
| | - Asish Pal
- Chemical Biology Unit, Institute of Nano Science and Technology (INST), Sector 81, Mohali, Punjab, 140306, India
| |
Collapse
|
12
|
Meyer N, Torrent J, Balme S. Characterizing Prion-Like Protein Aggregation: Emerging Nanopore-Based Approaches. SMALL METHODS 2024; 8:e2400058. [PMID: 38644684 PMCID: PMC11672191 DOI: 10.1002/smtd.202400058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/10/2024] [Indexed: 04/23/2024]
Abstract
Prion-like protein aggregation is characteristic of numerous neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. This process involves the formation of aggregates ranging from small and potentially neurotoxic oligomers to highly structured self-propagating amyloid fibrils. Various approaches are used to study protein aggregation, but they do not always provide continuous information on the polymorphic, transient, and heterogeneous species formed. This review provides an updated state-of-the-art approach to the detection and characterization of a wide range of protein aggregates using nanopore technology. For each type of nanopore, biological, solid-state polymer, and nanopipette, discuss the main achievements for the detection of protein aggregates as well as the significant contributions to the understanding of protein aggregation and diagnostics.
Collapse
Affiliation(s)
- Nathan Meyer
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonCedex 5Montpellier34095France
- INMUniversity of MontpellierINSERMMontpellier34095France
| | - Joan Torrent
- INMUniversity of MontpellierINSERMMontpellier34095France
| | - Sébastien Balme
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonCedex 5Montpellier34095France
| |
Collapse
|
13
|
Dey A, Naranjo E, Saha R, Zhang S, Nair MN, Li TD, Chen X, Ulijn RV. Water-Vapor Responsive Metallo-Peptide Nanofibers. Angew Chem Int Ed Engl 2024; 63:e202409391. [PMID: 39137360 DOI: 10.1002/anie.202409391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/15/2024]
Abstract
Short peptides are versatile molecules for the construction of supramolecular materials. Most reported peptide materials are hydrophobic, stiff, and show limited response to environmental conditions in the solid-state. Herein, we describe a design strategy for minimalistic supramolecular metallo-peptide nanofibers that, depending on their sequence, change stiffness, or reversibly assemble in the solid-state, in response to changes in relative humidity (RH). We tested a series of histidine (H) containing dipeptides with varying hydrophobicity, XH, where X is G, A, L, Y (glycine, alanine, leucine, and tyrosine). The one-dimensional fiber formation is supported by metal coordination and dynamic H-bonds. Solvent conditions were identified where GH/Zn and AH/Zn formed gels that upon air-drying gave rise to nanofibers. Upon exposure of the nanofiber networks to increasing RH, a reduction in stiffness was observed with GH/Zn fibers reversibly (dis-)assembled at 60-70 % RH driven by a rebalancing of hydrogen bonding interactions between peptides and water. When these metallo-peptide nanofibers were deposited on the surface of polyimide films and exposed to varying RH, peptide/water-vapor interactions in the solid-state mechanically transferred to the polymer film, leading to the rapid and reversible folding-unfolding of the films, thus demonstrating RH-responsive actuation.
Collapse
Affiliation(s)
- Avishek Dey
- Nanoscience Initiative at Advanced Science Research Center of the Graduate Center of the City University of New York, New York, New York, 10031, USA
| | - Elma Naranjo
- Nanoscience Initiative at Advanced Science Research Center of the Graduate Center of the City University of New York, New York, New York, 10031, USA
- Department of Chemical Engineering, The City College of New York, 275 Convent Ave, New York, NY 10031, USA
| | - Ranajit Saha
- Department of Chemistry, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, 736101, India
| | - Sheng Zhang
- Nanoscience Initiative at Advanced Science Research Center of the Graduate Center of the City University of New York, New York, New York, 10031, USA
| | - Maya Narayanan Nair
- Nanoscience Initiative at Advanced Science Research Center of the Graduate Center of the City University of New York, New York, New York, 10031, USA
| | - Tai-De Li
- Nanoscience Initiative at Advanced Science Research Center of the Graduate Center of the City University of New York, New York, New York, 10031, USA
- Department of Physics, City College of New York of City, University of New York, New York, NY 10031, USA
| | - Xi Chen
- Nanoscience Initiative at Advanced Science Research Center of the Graduate Center of the City University of New York, New York, New York, 10031, USA
- Department of Chemical Engineering, The City College of New York, 275 Convent Ave, New York, NY 10031, USA
- Ph.D. Program in Physics, The Graduate Center of the City University of New York, 365 5th Ave, New York, NY 10016, USA
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, 365 5th Ave, New York, NY 10016, USA
| | - Rein V Ulijn
- Nanoscience Initiative at Advanced Science Research Center of the Graduate Center of the City University of New York, New York, New York, 10031, USA
- Department of Chemistry Hunter College, City University of New York, New York, New York, 10065, USA
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, 365 5th Ave, New York, NY 10016, USA
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, 365 5th Ave, New York, NY 10016, USA
| |
Collapse
|
14
|
Seychell RM, El Saghir A, Vassallo N. Modulation of Biological Membranes Using Small-Molecule Compounds to Counter Toxicity Caused by Amyloidogenic Proteins. MEMBRANES 2024; 14:231. [PMID: 39590617 PMCID: PMC11596372 DOI: 10.3390/membranes14110231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
The transition of peptides or proteins along a misfolding continuum from soluble functional states to pathological aggregates, to ultimately deposit as amyloid fibrils, is a process that underlies an expanding group of human diseases-collectively known as protein-misfolding disorders (PMDs). These include common and debilitating conditions, such as Alzheimer's disease, Parkinson's disease, and type-2 diabetes. Compelling evidence has emerged that the complex interplay between the misfolded proteins and biological membranes is a key determinant of the pathogenic mechanisms by which harmful amyloid entities are formed and exert their cytotoxicity. Most efforts thus far to develop disease-modifying treatments for PMDs have largely focused on anti-aggregation strategies: to neutralise, or prevent the formation of, toxic amyloid species. Herein, we review the critical role of the phospholipid membrane in mediating and enabling amyloid pathogenicity. We consequently propose that the development of small molecules, which have the potential to uniquely modify the physicochemical properties of the membrane and make it more resilient against damage by misfolded proteins, could provide a novel therapeutic approach in PMDs. By way of an example, natural compounds shown to intercalate into lipid bilayers and inhibit amyloid-lipid interactions, such as the aminosterols, squalamine and trodusquamine, cholesterol, ubiquinone, and select polyphenols, are discussed. Such a strategy would provide a novel approach to counter a wide range of toxic biomolecules implicit in numerous human amyloid pathologies.
Collapse
Affiliation(s)
- Raina Marie Seychell
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Adam El Saghir
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, MSD 2080 Msida, Malta
| |
Collapse
|
15
|
Di Lorenzo D. Tau Protein and Tauopathies: Exploring Tau Protein-Protein and Microtubule Interactions, Cross-Interactions and Therapeutic Strategies. ChemMedChem 2024; 19:e202400180. [PMID: 39031682 DOI: 10.1002/cmdc.202400180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/22/2024]
Abstract
Tau, a microtubule-associated protein (MAP), is essential to maintaining neuronal stability and function in the healthy brain. However, aberrant modifications and pathological aggregations of Tau are implicated in various neurodegenerative disorders, collectively known as tauopathies. The most common Tauopathy is Alzheimer's Disease (AD) counting nowadays more than 60 million patients worldwide. This comprehensive review delves into the multifaceted realm of Tau protein, puzzling out its intricate involvement in both physiological and pathological roles. Emphasis is put on Tau Protein-Protein Interactions (PPIs), depicting its interaction with tubulin, microtubules and its cross-interaction with other proteins such as Aβ1-42, α-synuclein, and the chaperone machinery. In the realm of therapeutic strategies, an overview of diverse possibilities is presented with their relative clinical progresses. The focus is mostly addressed to Tau protein aggregation inhibitors including recent small molecules, short peptides and peptidomimetics with specific focus on compounds that showed a double anti aggregative activity on both Tau protein and Aβ amyloid peptide. This review amalgamates current knowledge on Tau protein and evolving therapeutic strategies, providing a comprehensive resource for researchers seeking to deepen their understanding of the Tau protein and for scientists involved in the development of new peptide-based anti-aggregative Tau compounds.
Collapse
Affiliation(s)
- Davide Di Lorenzo
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, D-33615, Bielefeld, Germany
| |
Collapse
|
16
|
Gonçalves PB, Sodero ACR, Cordeiro Y. Natural products targeting amyloid-β oligomer neurotoxicity in Alzheimer's disease. Eur J Med Chem 2024; 276:116684. [PMID: 39032401 DOI: 10.1016/j.ejmech.2024.116684] [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: 06/06/2024] [Revised: 07/07/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Alzheimer's disease (AD) constitutes a major global health issue, characterized by progressive neurodegeneration and cognitive impairment, for which no curative treatment is currently available. Current therapeutic approaches are focused on symptom management, highlighting the critical need for disease-modifying therapy. The hallmark pathology of AD involves the aggregation and accumulation of amyloid-β (Aβ) peptides in the brain. Consequently, drug discovery efforts in recent decades have centered on the Aβ aggregation cascade, which includes the transition of monomeric Aβ peptides into toxic oligomers and, ultimately, mature fibrils. Historically, anti-Aβ strategies focused on the clearance of amyloid fibrils using monoclonal antibodies. However, substantial evidence has highlighted the critical role of Aβ oligomers (AβOs) in AD pathogenesis. Soluble AβOs are now recognized as more toxic than fibrils, directly contributing to synaptic impairment, neuronal damage, and the onset of AD. Targeting AβOs has emerged as a promising therapeutic approach to mitigate cognitive decline in AD. Natural products (NPs) have demonstrated promise against AβO neurotoxicity through various mechanisms, including preventing AβO formation, enhancing clearance mechanisms, or converting AβOs into non-toxic species. Understanding the mechanisms by which anti-AβO NPs operate is useful for developing disease-modifying treatments for AD. In this review, we explore the role of NPs in mitigating AβO neurotoxicity for AD drug discovery, summarizing key evidence from biophysical methods, cellular assays, and animal models. By discussing how NPs modulate AβO neurotoxicity across various experimental systems, we aim to provide valuable insights into novel therapeutic strategies targeting AβOs in AD.
Collapse
Affiliation(s)
| | | | - Yraima Cordeiro
- Faculty of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, 21949-900, Brazil
| |
Collapse
|
17
|
Lin PH, Wu GW, Lin YH, Huang JR, Jeng US, Liu WM, Huang JR. TDP-43 Amyloid Fibril Formation via Phase Separation-Related and -Unrelated Pathways. ACS Chem Neurosci 2024; 15. [PMID: 39358890 PMCID: PMC11488477 DOI: 10.1021/acschemneuro.4c00503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024] Open
Abstract
Intrinsically disordered regions (IDRs) in proteins can undergo liquid-liquid phase separation (LLPS) for functional assembly, but this increases the chance of forming disease-associated amyloid fibrils. Not all amyloid fibrils form through LLPS however, and the importance of LLPS relative to other pathways in fibril formation remains unclear. We investigated this question in TDP-43, a motor neuron disease and dementia-causing protein that undergoes LLPS, using thioflavin T (ThT) fluorescence, NMR, transmission electron microscopy (TEM), and wide-angle X-ray scattering (WAXS) experiments. Using a fluorescence probe modified from ThT strategically designed for targeting protein assembly rather than β-sheets and supported by TEM images, we propose that the biphasic ThT signals observed under LLPS-favoring conditions are due to the presence of amorphous aggregates. These aggregates represent an intermediate state that diverges from the direct pathway to β-sheet-dominant fibrils. Under non-LLPS conditions in contrast (at low pH or at physiological conditions in a construct with key LLPS residues removed), the protein forms a hydrogel. Real-time WAXS data, ThT signals, and TEM images collectively demonstrate that the gelation process circumvents LLPS and yet still results in the formation of fibril-like structural networks. We suggest that the IDR of TDP-43 forms disease-causing amyloid fibrils regardless of the formation pathway. Our findings shed light on why both LLPS-promoting and LLPS-inhibiting mutants are found in TDP-43-related diseases.
Collapse
Affiliation(s)
- Pin-Han Lin
- Institute
of Biochemistry and Molecular Biology, National
Yang Ming Chiao Tung University, No. 155 Section 2, Li-nong Street, Taipei 11221, Taiwan
| | - Guan-Wei Wu
- Institute
of Biochemistry and Molecular Biology, National
Yang Ming Chiao Tung University, No. 155 Section 2, Li-nong Street, Taipei 11221, Taiwan
| | - Yu-Hao Lin
- Institute
of Biochemistry and Molecular Biology, National
Yang Ming Chiao Tung University, No. 155 Section 2, Li-nong Street, Taipei 11221, Taiwan
| | - Jing-Rou Huang
- Institute
of Biochemistry and Molecular Biology, National
Yang Ming Chiao Tung University, No. 155 Section 2, Li-nong Street, Taipei 11221, Taiwan
| | - U-Ser Jeng
- National
Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan
| | - Wei-Min Liu
- Department
of Chemistry, Fu Jen Catholic University, No.510, Zhongzheng Rd., New Taipei City 24205, Taiwan
| | - Jie-rong Huang
- Institute
of Biochemistry and Molecular Biology, National
Yang Ming Chiao Tung University, No. 155 Section 2, Li-nong Street, Taipei 11221, Taiwan
- Institute
of Biomedical Informatics, National Yang
Ming Chiao Tung University, No. 155 Section 2, Li-nong Street, Taipei 11221, Taiwan
- Department
of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, No. 155 Section 2, Li-nong Street, Taipei 11221, Taiwan
| |
Collapse
|
18
|
Meyer N, Arroyo N, Roustan L, Janot J, Charles‐Achille S, Torrent J, Picaud F, Balme S. Secondary Nucleation of Aβ Revealed by Single-Molecule and Computational Approaches. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404916. [PMID: 39159070 PMCID: PMC11497034 DOI: 10.1002/advs.202404916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/07/2024] [Indexed: 08/21/2024]
Abstract
Understanding the mechanisms underlying amyloid-β (Aβ) aggregation is pivotal in the context of Alzheimer's disease. This study aims to elucidate the secondary nucleation process of Aβ42 peptides by combining experimental and computational methods. Using a newly developed nanopipette-based amyloid seeding and translocation assay, confocal fluorescence spectroscopy, and molecular dynamics simulations, the influence of the seed properties on Aβ aggregation is investigated. Both fragmented and unfragmented seeds played distinct roles in the formation of oligomers, with fragmented seeds facilitating the formation of larger aggregates early in the incubation phase. The results show that secondary nucleation leads to the formation of oligomers of various sizes and structures as well as larger fibrils structured in β-sheets. From these findings a mechanism of secondary nucleation involving two types of aggregate populations, one released and one growing on the mother fiber is proposed.
Collapse
Affiliation(s)
- Nathan Meyer
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonMontpellier34095France
| | - Nicolas Arroyo
- UR SINERGIESUniversity of Besançon16 route de GrayBesançon25000France
| | - Lois Roustan
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonMontpellier34095France
| | - Jean‐Marc Janot
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonMontpellier34095France
| | - Saly Charles‐Achille
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonMontpellier34095France
| | - Joan Torrent
- INMUniversity of MontpellierINSERMMontpellier34090France
| | - Fabien Picaud
- UR SINERGIESUniversity of Besançon16 route de GrayBesançon25000France
| | - Sébastien Balme
- Institut Européen des MembranesUMR5635 University of Montpellier ENCSM CNRSPlace Eugène BataillonMontpellier34095France
| |
Collapse
|
19
|
Dabas A, Goyal B. Structural Reorganization Mechanism of the Aβ 42 Fibril Mediated by N-Substituted Oligopyrrolamide ADH-353. ACS Chem Neurosci 2024; 15:3136-3151. [PMID: 39158263 DOI: 10.1021/acschemneuro.4c00253] [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] [Indexed: 08/20/2024] Open
Abstract
The inhibition of amyloid-β (Aβ) fibrillation and clearance of Aβ aggregates have emerged as a potential pharmacological strategy to alleviate Aβ aggregate-induced neurotoxicity in Alzheimer's disease (AD). Maity et al. shortlisted ADH-353 from a small library of positively charged N-substituted oligopyrrolamides for its notable ability to inhibit Aβ fibrillation, disintegrate intracellular cytotoxic Aβ oligomers, and alleviate Aβ-induced cytotoxicity in the SH-SY5Y and N2a cells. However, the molecular mechanism through which ADH-353 interacts with the Aβ42 fibrils, leading to their disruption and subsequent clearance, remains unclear. Thus, a detailed molecular mechanism underlying the disruption of neurotoxic Aβ42 fibrils (PDB ID 2NAO) by ADH-353 has been illuminated in this work using molecular dynamics simulations. Interestingly, conformational snapshots during simulation depicted the shortening and disappearance of β-strands and the emergence of a helix conformation, indicating a loss of the well-organized β-sheet-rich structure of the disease-relevant Aβ42 fibril on the incorporation of ADH-353. ADH-353 binds strongly to the Aβ42 fibril (ΔGbinding= -142.91 ± 1.61 kcal/mol) with a notable contribution from the electrostatic interactions between positively charged N-propylamine side chains of ADH-353 with the glutamic (Glu3, Glu11, and Glu22) and aspartic (Asp7 and Asp23) acid residues of the Aβ42 fibril. This aligns well with heteronuclear single quantum coherence NMR studies, which depict that the binding of ADH-353 with the Aβ peptide is driven by electrostatic and hydrophobic contacts. Furthermore, a noteworthy decrease in the binding affinity of Aβ42 fibril chains on the incorporation of ADH-353 indicates the weakening of interchain interactions leading to the disruption of the double-horseshoe conformation of the Aβ42 fibril. The illumination of key interactions responsible for the destabilization of the Aβ42 fibril by ADH-353 in this work will greatly aid in designing new chemical scaffolds with enhanced efficacy for the clearance of Aβ aggregates in AD.
Collapse
Affiliation(s)
- Arushi Dabas
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| | - Bhupesh Goyal
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| |
Collapse
|
20
|
Krawczuk D, Groblewska M, Mroczko J, Winkel I, Mroczko B. The Role of α-Synuclein in Etiology of Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9197. [PMID: 39273146 PMCID: PMC11395629 DOI: 10.3390/ijms25179197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
A presynaptic protein called α-synuclein plays a crucial role in synaptic function and neurotransmitter release. However, its misfolding and aggregation have been implicated in a variety of neurodegenerative diseases, particularly Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. Emerging evidence suggests that α-synuclein interacts with various cellular pathways, including mitochondrial dysfunction, oxidative stress, and neuroinflammation, which contributes to neuronal cell death. Moreover, α-synuclein has been involved in the propagation of neurodegenerative processes through prion-like mechanisms, where misfolded proteins induce similar conformational changes in neighboring neurons. Understanding the multifaced roles of α-synuclein in neurodegeneration not only aids in acquiring more knowledge about the pathophysiology of these diseases but also highlights potential biomarkers and therapeutic targets for intervention in alpha-synucleinopathies. In this review, we provide a summary of the mechanisms by which α-synuclein contributes to neurodegenerative processes, focusing on its misfolding, oligomerization, and the formation of insoluble fibrils that form characteristic Lewy bodies. Furthermore, we compare the potential value of α-synuclein species in diagnosing and differentiating selected neurodegenerative diseases.
Collapse
Affiliation(s)
- Daria Krawczuk
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland; (D.K.); (J.M.)
| | - Magdalena Groblewska
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
| | - Jan Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland; (D.K.); (J.M.)
| | - Izabela Winkel
- Dementia Disorders Centre, Medical University of Wroclaw, 50-425 Ścinawa, Poland;
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-089 Białystok, Poland; (D.K.); (J.M.)
- Department of Biochemical Diagnostics, University Hospital in Białystok, 15-269 Białystok, Poland;
| |
Collapse
|
21
|
Xu CK, Meisl G, Andrzejewska EA, Krainer G, Dear AJ, Castellana-Cruz M, Turi S, Edu IA, Vivacqua G, Jacquat RPB, Arter WE, Spillantini MG, Vendruscolo M, Linse S, Knowles TPJ. α-Synuclein oligomers form by secondary nucleation. Nat Commun 2024; 15:7083. [PMID: 39153989 PMCID: PMC11330488 DOI: 10.1038/s41467-024-50692-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 07/19/2024] [Indexed: 08/19/2024] Open
Abstract
Oligomeric species arising during the aggregation of α-synuclein are implicated as a major source of toxicity in Parkinson's disease, and thus a major potential drug target. However, both their mechanism of formation and role in aggregation are largely unresolved. Here we show that, at physiological pH and in the absence of lipid membranes, α-synuclein aggregates form by secondary nucleation, rather than simple primary nucleation, and that this process is enhanced by agitation. Moreover, using a combination of single molecule and bulk level techniques, we identify secondary nucleation on the surfaces of existing fibrils, rather than formation directly from monomers, as the dominant source of oligomers. Our results highlight secondary nucleation as not only the key source of oligomers, but also the main mechanism of aggregate formation, and show that these processes take place under conditions which recapitulate the neutral pH and ionic strength of the cytosol.
Collapse
Affiliation(s)
- Catherine K Xu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Max Planck Institute for the Science of Light, Erlangen, Germany
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Ewa A Andrzejewska
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Georg Krainer
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Institute of Molecular Biosciences (IMB), University of Graz, Graz, Austria
| | - Alexander J Dear
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Marta Castellana-Cruz
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Soma Turi
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Irina A Edu
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Giorgio Vivacqua
- Integrated Research Center (PRAAB), Campus Biomedico University of Rome, Rome, Italy
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Raphaël P B Jacquat
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - William E Arter
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | | | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Sara Linse
- Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
22
|
Li J, Zhang Y, Dong J, Li D, Ba X, Wang S. Dissimilar effects of the hydrophilic carbon dots on the amyloid aggregation of two model proteins and the mechanism discussion. J Mol Recognit 2024; 37:e3085. [PMID: 38599335 DOI: 10.1002/jmr.3085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/04/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024]
Abstract
Many proteins could aggregate into amyloid fibrils under certain conditions. However, the aggregation process and morphology of the fibrils may be significantly different because of the distinct protein structure. In this article, the hydrophilic carbon dots (Lys-CA-CDs) were prepared using lysine (Lys) and citric acid (CA) as reactant under the assistance of a microwave. The dissimilar modulation effect of Lys-CA-CDs on the aggregation process of distinct structure protein was further investigated, where bovine serum albumin (BSA) and hen egg white lysozyme (HEWL) were chosen as model proteins. All results showed that Lys-CA-CDs displayed the contrary influence on the aggregation process of BSA and HEWL. Lys-CA-CDs could induce BSA to aggregate into more wormlike fibrils and inhibit the aggregation of HEWL into hair-like fibrils. The influence on the aggregation process of BSA may be assigned to the increased concentration of BSA around the Lys-CA-CDs caused by their interaction. However, inserting of Lys-CA-CDs into the inner structure of HEWL led to the change of protein secondary structure. The change of secondary structure further made it difficult for HEWL to aggregate into fibrils and Lys-CA-CDs showed the inhibition effect on HEWL aggregation.
Collapse
Affiliation(s)
- Jie Li
- College of Chemistry and Materials Science, Hebei University, Baoding, P. R. China
| | - Yuangong Zhang
- School of Basic Medical Sciences, Hebei University, Baoding, P. R. China
| | - Jiawei Dong
- College of Chemistry and Materials Science, Hebei University, Baoding, P. R. China
| | - Dexin Li
- College of Chemistry and Materials Science, Hebei University, Baoding, P. R. China
| | - Xinwu Ba
- College of Chemistry and Materials Science, Hebei University, Baoding, P. R. China
| | - Sujuan Wang
- College of Chemistry and Materials Science, Hebei University, Baoding, P. R. China
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Hebei University, Baoding, P. R. China
| |
Collapse
|
23
|
Keith A, Brichtová EP, Barber JG, Wales DJ, Jackson SE, Röder K. Energy Landscapes and Structural Ensembles of Glucagon-like Peptide-1 Monomers. J Phys Chem B 2024; 128:5601-5611. [PMID: 38831581 PMCID: PMC11182347 DOI: 10.1021/acs.jpcb.4c01794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
While GLP-1 and its analogues are important pharmaceutical agents in the treatment of type 2 diabetes and obesity, their susceptibility to aggregate into amyloid fibrils poses a significant safety issue. Many factors may contribute to the aggregation propensity, including pH. While it is known that the monomeric structure of GLP-1 has a strong impact on primary nucleation, probing its diverse structural ensemble is challenging. Here, we investigated the monomer structural ensembles at pH 3, 4, and 7.5 using state-of-the-art computational methods in combination with experimental data. We found significant stabilization of β-strand structures and destabilization of helical structures at lower pH, correlating with observed aggregation lag times, which are lower under these conditions. We further identified helical defects at pH 4, which led to the fastest observed aggregation, in agreement with our far-UV circular dichroism data. The detailed atomistic structures that result from the computational studies help to rationalize the experimental results on the aggregation propensity of GLP-1. This work provides a new insight into the pH-dependence of monomeric structural ensembles of GLP-1 and connects them to experimental observations.
Collapse
Affiliation(s)
- Alasdair
D. Keith
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
- Now:
Department of Biochemistry, School of Medicine, Emory University, 1510 Clifton Rd NE, Atlanta, Georgia 30322, United States
| | - Eva Přáda Brichtová
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
- Now:
Institute of Chemical, Environmental and Bioscience Engineering, Technische Universität Wien, Gumpendorferstr. 1A, Vienna 1060, Austria
| | - Jack G. Barber
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - David J. Wales
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Sophie E. Jackson
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Konstantin Röder
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
- Now:
Randall Centre for Cell & Molecular Biophysics, King’s College London, Great Maze Pond, London SE1 1UL, U.K.
| |
Collapse
|
24
|
Kaur G, Mankoo OK, Kaur A, Goyal D, Goyal B. Insights into the baicalein-induced destabilization of LS-shaped Aβ 42 protofibrils using computer simulations. Phys Chem Chem Phys 2024; 26:16674-16686. [PMID: 38809059 DOI: 10.1039/d3cp06006c] [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: 05/30/2024]
Abstract
Amyloid-β (Aβ) peptides aggregate spontaneously into various aggregating species comprising oligomers, protofibrils, and mature fibrils in Alzheimer's disease (AD). Disrupting β-sheet rich neurotoxic smaller soluble Aβ42 oligomers formed at early stages is considered a potent strategy to interfere with AD pathology. Previous experiments have demonstrated the inhibition of the early stages of Aβ aggregation by baicalein; however, the molecular mechanism behind inhibition remains largely unknown. Thus, in this work, molecular dynamics (MD) simulations have been employed to illuminate the molecular mechanism of baicalein-induced destabilization of preformed Aβ42 protofibrils. Baicalein binds to chain A of the Aβ42 protofibril through hydrogen bonds, π-π interactions, and hydrophobic contacts with the central hydrophobic core (CHC) residues of the Aβ42 protofibril. The binding of baicalein to the CHC region of the Aβ42 protofibril resulted in the elongation of the kink angle and disruption of K28-A42 salt bridges, which resulted in the distortion of the protofibril structure. Importantly, the β-sheet content was notably reduced in Aβ42 protofibrils upon incorporation of baicalein with a concomitant increase in the coil content, which is consistent with ThT fluorescence and AFM images depicting disaggregation of pre-existing Aβ42 fibrils on the incorporation of baicalein. Remarkably, the interchain binding affinity in Aβ42 protofibrils was notably reduced in the presence of baicalein leading to distortion in the overall structure, which agrees with the structural stability analyses and conformational snapshots. This work sheds light on the molecular mechanism of baicalein in disrupting the Aβ42 protofibril structure, which will be beneficial to the design of therapeutic candidates against disrupting β-sheet rich neurotoxic Aβ42 oligomers in AD.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib-140406, Punjab, India
| | - Opinder Kaur Mankoo
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib-140406, Punjab, India
| | - Anupamjeet Kaur
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib-140406, Punjab, India
| | - Deepti Goyal
- Department of Chemistry, DAV College, Sector 10, Chandigarh-160011, India.
| | - Bhupesh Goyal
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala-147004, Punjab, India.
| |
Collapse
|
25
|
Dear A, Thacker D, Wennmalm S, Ortigosa-Pascual L, Andrzejewska EA, Meisl G, Linse S, Knowles TPJ. Aβ Oligomer Dissociation Is Catalyzed by Fibril Surfaces. ACS Chem Neurosci 2024; 15:2296-2307. [PMID: 38785363 PMCID: PMC11157482 DOI: 10.1021/acschemneuro.4c00127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Oligomeric assemblies consisting of only a few protein subunits are key species in the cytotoxicity of neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. Their lifetime in solution and abundance, governed by the balance of their sources and sinks, are thus important determinants of disease. While significant advances have been made in elucidating the processes that govern oligomer production, the mechanisms behind their dissociation are still poorly understood. Here, we use chemical kinetic modeling to determine the fate of oligomers formed in vitro and discuss the implications for their abundance in vivo. We discover that oligomeric species formed predominantly on fibril surfaces, a broad class which includes the bulk of oligomers formed by the key Alzheimer's disease-associated Aβ peptides, also dissociate overwhelmingly on fibril surfaces, not in solution as had previously been assumed. We monitor this "secondary nucleation in reverse" by measuring the dissociation of Aβ42 oligomers in the presence and absence of fibrils via two distinct experimental methods. Our findings imply that drugs that bind fibril surfaces to inhibit oligomer formation may also inhibit their dissociation, with important implications for rational design of therapeutic strategies for Alzheimer's and other amyloid diseases.
Collapse
Affiliation(s)
- Alexander
J. Dear
- Biochemistry
and Structural Biology, Lund University, Lund 221 00, Sweden
- Centre
for Misfolding Diseases Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Dev Thacker
- Biochemistry
and Structural Biology, Lund University, Lund 221 00, Sweden
| | - Stefan Wennmalm
- Department
of Applied Physics, Biophysics Group, SciLifeLab, Royal Institute of Technology-KTH, Solna 171 65, Sweden
| | | | - Ewa A. Andrzejewska
- Centre
for Misfolding Diseases Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Georg Meisl
- Centre
for Misfolding Diseases Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Sara Linse
- Biochemistry
and Structural Biology, Lund University, Lund 221 00, Sweden
| | - Tuomas P. J. Knowles
- Centre
for Misfolding Diseases Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
- Cavendish
Laboratory, University of Cambridge, J J Thomson Avenue, Cambridge CB3 0HE, U.K.
| |
Collapse
|
26
|
Mukherjee S, Poudyal M, Dave K, Kadu P, Maji SK. Protein misfolding and amyloid nucleation through liquid-liquid phase separation. Chem Soc Rev 2024; 53:4976-5013. [PMID: 38597222 DOI: 10.1039/d3cs01065a] [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: 04/11/2024]
Abstract
Liquid-liquid phase separation (LLPS) is an emerging phenomenon in cell physiology and diseases. The weak multivalent interaction prerequisite for LLPS is believed to be facilitated through intrinsically disordered regions, which are prevalent in neurodegenerative disease-associated proteins. These aggregation-prone proteins also exhibit an inherent property for phase separation, resulting in protein-rich liquid-like droplets. The very high local protein concentration in the water-deficient confined microenvironment not only drives the viscoelastic transition from the liquid to solid-like state but also most often nucleate amyloid fibril formation. Indeed, protein misfolding, oligomerization, and amyloid aggregation are observed to be initiated from the LLPS of various neurodegeneration-related proteins. Moreover, in these cases, neurodegeneration-promoting genetic and environmental factors play a direct role in amyloid aggregation preceded by the phase separation. These cumulative recent observations ignite the possibility of LLPS being a prominent nucleation mechanism associated with aberrant protein aggregation. The present review elaborates on the nucleation mechanism of the amyloid aggregation pathway and the possible early molecular events associated with amyloid-related protein phase separation. It also summarizes the recent advancement in understanding the aberrant phase transition of major proteins contributing to neurodegeneration focusing on the common disease-associated factors. Overall, this review proposes a generic LLPS-mediated multistep nucleation mechanism for amyloid aggregation and its implication in neurodegeneration.
Collapse
Affiliation(s)
- Semanti Mukherjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Manisha Poudyal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Kritika Dave
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| | - Samir K Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
27
|
Iscen A, Kaygisiz K, Synatschke CV, Weil T, Kremer K. Multiscale Simulations of Self-Assembling Peptides: Surface and Core Hydrophobicity Determine Fibril Stability and Amyloid Aggregation. Biomacromolecules 2024; 25:3063-3075. [PMID: 38652055 PMCID: PMC11094720 DOI: 10.1021/acs.biomac.4c00151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024]
Abstract
Assemblies of peptides and proteins through specific intermolecular interactions set the basis for macroscopic materials found in nature. Peptides provide easily tunable hydrogen-bonding interactions, which can lead to the formation of ordered structures such as highly stable β-sheets that can form amyloid-like supramolecular peptide nanofibrils (PNFs). PNFs are of special interest, as they could be considered as mimics of various fibrillar structures found in nature. In their ability to serve as supramolecular scaffolds, they could mimic certain features of the extracellular matrix to provide stability, interact with pathogens such as virions, and transduce signals between the outside and inside of cells. Many PNFs have been reported that reveal rich bioactivities. PNFs supporting neuronal cell growth or lentiviral gene transduction have been studied systematically, and their material properties were correlated to bioactivities. However, the impact of the structure of PNFs, their dynamics, and stabilities on their unique functions is still elusive. Herein, we provide a microscopic view of the self-assembled PNFs to unravel how the amino acid sequence of self-assembling peptides affects their secondary structure and dynamic properties of the peptides within supramolecular fibrils. Based on sequence truncation, amino acid substitution, and sequence reordering, we demonstrate that peptide-peptide aggregation propensity is critical to form bioactive β-sheet-rich structures. In contrast to previous studies, a very high peptide aggregation propensity reduces bioactivity due to intermolecular misalignment and instabilities that emerge when fibrils are in close proximity to other fibrils in solution. Our multiscale simulation approach correlates changes in biological activity back to single amino acid modifications. Understanding these relationships could lead to future material discoveries where the molecular sequence predictably determines the macroscopic properties and biological activity. In addition, our studies may provide new insights into naturally occurring amyloid fibrils in neurodegenerative diseases.
Collapse
Affiliation(s)
- Aysenur Iscen
- Department
of Polymer Theory, Max Planck Institute
for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Kübra Kaygisiz
- Department
of Synthesis of Macromolecules, Max Planck
Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Christopher V. Synatschke
- Department
of Synthesis of Macromolecules, Max Planck
Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Tanja Weil
- Department
of Synthesis of Macromolecules, Max Planck
Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Kurt Kremer
- Department
of Polymer Theory, Max Planck Institute
for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
28
|
Patra S, Chandrabhas S, Dhiman S, George SJ. Controlled Supramolecular Polymerization via Bioinspired, Liquid-Liquid Phase Separation of Monomers. J Am Chem Soc 2024; 146:12577-12586. [PMID: 38683934 DOI: 10.1021/jacs.4c01377] [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: 05/02/2024]
Abstract
Dynamic supramolecular assemblies, driven by noncovalent interactions, pervade the biological realm. In the synthetic domain, their counterparts, supramolecular polymers, endowed with remarkable self-repair and adaptive traits, are often realized through bioinspired designs. Recently, controlled supramolecular polymerization strategies have emerged, drawing inspiration from protein self-assembly. A burgeoning area of research involves mimicking the liquid-liquid phase separation (LLPS) observed in proteins to create coacervate droplets and recognizing their significance in cellular organization and diverse functions. Herein, we introduce a novel perspective on synthetic coacervates, extending beyond their established role in synthetic biology as dynamic, membraneless phases to enable structural control in synthetic supramolecular polymers. Drawing parallels with the cooperative growth of amyloid fibrils through LLPS, we present metastable coacervate droplets as dormant monomer phases for controlled supramolecular polymerization. This is achieved via a π-conjugated monomer design that combines structural characteristics for both coacervation through its terminal ionic groups and one-dimensional growth via a π-conjugated core. This design leads to a unique temporal LLPS, resulting in a metastable coacervate phase, which subsequently undergoes one-dimensional growth via nucleation within the droplets. In-depth spectroscopic and microscopic characterization provides insights into the temporal evolution of disordered and ordered phases. Furthermore, to modulate the kinetics of liquid-to-solid transformation and to achieve precise control over the structural characteristics of the resulting supramolecular polymers, we invoke seeding in the droplets, showcasing living growth characteristics. Our work thus opens up new avenues in the exciting field of supramolecular polymerization, offering general design principles and controlled synthesis of precision self-assembled structures in confined environments.
Collapse
Affiliation(s)
- Satyajit Patra
- New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore 560064, India
| | - Sushmitha Chandrabhas
- New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore 560064, India
| | - Shikha Dhiman
- New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore 560064, India
| | - Subi J George
- New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore 560064, India
| |
Collapse
|
29
|
Agha MM, Aziziyan F, Uversky VN. Each big journey starts with a first step: Importance of oligomerization. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 206:111-141. [PMID: 38811079 DOI: 10.1016/bs.pmbts.2024.03.011] [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: 05/31/2024]
Abstract
Protein oligomers, widely found in nature, have significant physiological and pathological functions. They are classified into three groups based on their function and toxicity. Significant advancements are being achieved in the development of functional oligomers, with a focus on various applications and their engineering. The antimicrobial peptides oligomers play roles in death of bacterial and cancer cells. The predominant pathogenic species in neurodegenerative disorders, as shown by recent results, are amyloid oligomers, which are the main subject of this chapter. They are generated throughout the aggregation process, serving as both intermediates in the subsequent aggregation pathways and ultimate products. Some of them may possess potent cytotoxic properties and through diverse mechanisms cause cellular impairment, and ultimately, the death of cells and disease progression. Information regarding their structure, formation mechanism, and toxicity is limited due to their inherent instability and structural variability. This chapter aims to provide a concise overview of the current knowledge regarding amyloid oligomers.
Collapse
Affiliation(s)
- Mansoureh Mirza Agha
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Aziziyan
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vladimir N Uversky
- Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, Institute for Biological Instrumentation, Pushchino, Moscow, Russia; Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United Staes.
| |
Collapse
|
30
|
Meisl G. The thermodynamics of neurodegenerative disease. BIOPHYSICS REVIEWS 2024; 5:011303. [PMID: 38525484 PMCID: PMC10957229 DOI: 10.1063/5.0180899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/26/2024] [Indexed: 03/26/2024]
Abstract
The formation of protein aggregates in the brain is a central aspect of the pathology of many neurodegenerative diseases. This self-assembly of specific proteins into filamentous aggregates, or fibrils, is a fundamental biophysical process that can easily be reproduced in the test tube. However, it has been difficult to obtain a clear picture of how the biophysical insights thus obtained can be applied to the complex, multi-factorial diseases and what this means for therapeutic strategies. While new, disease-modifying therapies are now emerging, for the most devastating disorders, such as Alzheimer's and Parkinson's disease, they still fall well short of offering a cure, and few drug design approaches fully exploit the wealth of mechanistic insights that has been obtained in biophysical studies. Here, I attempt to provide a new perspective on the role of protein aggregation in disease, by phrasing the problem in terms of a system that, under constant energy consumption, attempts to maintain a healthy, aggregate-free state against the thermodynamic driving forces that inexorably push it toward pathological aggregation.
Collapse
Affiliation(s)
- Georg Meisl
- WaveBreak Therapeutics Ltd., Chemistry of Health, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
31
|
Marković M, Milošević J, Wang W, Cao Y. Passive Immunotherapies Targeting Amyloid- β in Alzheimer's Disease: A Quantitative Systems Pharmacology Perspective. Mol Pharmacol 2023; 105:1-13. [PMID: 37907353 DOI: 10.1124/molpharm.123.000726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 11/02/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by amyloid-β (Aβ) protein accumulation in the brain. Passive immunotherapies using monoclonal antibodies for targeting Aβ have shown promise for AD treatment. Indeed, recent US Food and Drug Administration approval of aducanumab and lecanemab, alongside positive donanemab Phase III results demonstrated clinical efficacy after decades of failed clinical trials for AD. However, the pharmacological basis distinguishing clinically effective from ineffective therapies remains unclear, impeding development of potent therapeutics. This study aimed to provide a quantitative perspective for effectively targeting Aβ with antibodies. We first reviewed the contradicting results associated with the amyloid hypothesis and the pharmacological basis of Aβ immunotherapy. Subsequently, we developed a quantitative systems pharmacology (QSP) model that describes the non-linear progression of Aβ pathology and the pharmacologic actions of the Aβ-targeting antibodies. Using the QSP model, we analyzed various scenarios for effective passive immunotherapy for AD. The model revealed that binding exclusively to the Aβ monomer has minimal effect on Aβ aggregation and plaque reduction, making the antibody affinity toward Aβ monomer unwanted, as it could become a distractive mechanism for plaque reduction. Neither early intervention, high brain penetration, nor increased dose could yield significant improvement of clinical efficacy for antibodies targeting solely monomers. Antibodies that bind all Aβ species but lack effector function exhibited moderate effects in plaque reduction. Our model highlights the importance of binding aggregate Aβ species and incorporating effector functions for efficient and early plaque reduction, guiding the development of more effective therapies for this devastating disease. SIGNIFICANCE STATEMENT: Despite previous unsuccessful attempts spanning several decades, passive immunotherapies utilizing monoclonal antibodies for targeting amyloid-beta (Aβ) have demonstrated promise with two recent FDA approvals. However, the pharmacological basis that differentiates clinically effective therapies from ineffective ones remains elusive. Our study offers a quantitative systems pharmacology perspective, emphasizing the significance of selectively targeting specific Aβ species and importance of antibody effector functions. This perspective sheds light on the development of more effective therapies for this devastating disease.
Collapse
Affiliation(s)
- Milica Marković
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy (M.M., Y.C.) and Lineberger Comprehensive Cancer Center, School of Medicine (Y.C.), University of North Carolina at Chapel Hill, North Carolina; Department of Biochemistry (J.M.), University of Belgrade, Faculty of Chemistry, Belgrade, Serbia; and Clinical Pharmacology and Pharmacometrics, Janssen Research & Development (W.W.), LLC, Spring House, Pennsylvania
| | - Jelica Milošević
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy (M.M., Y.C.) and Lineberger Comprehensive Cancer Center, School of Medicine (Y.C.), University of North Carolina at Chapel Hill, North Carolina; Department of Biochemistry (J.M.), University of Belgrade, Faculty of Chemistry, Belgrade, Serbia; and Clinical Pharmacology and Pharmacometrics, Janssen Research & Development (W.W.), LLC, Spring House, Pennsylvania
| | - Weirong Wang
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy (M.M., Y.C.) and Lineberger Comprehensive Cancer Center, School of Medicine (Y.C.), University of North Carolina at Chapel Hill, North Carolina; Department of Biochemistry (J.M.), University of Belgrade, Faculty of Chemistry, Belgrade, Serbia; and Clinical Pharmacology and Pharmacometrics, Janssen Research & Development (W.W.), LLC, Spring House, Pennsylvania
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy (M.M., Y.C.) and Lineberger Comprehensive Cancer Center, School of Medicine (Y.C.), University of North Carolina at Chapel Hill, North Carolina; Department of Biochemistry (J.M.), University of Belgrade, Faculty of Chemistry, Belgrade, Serbia; and Clinical Pharmacology and Pharmacometrics, Janssen Research & Development (W.W.), LLC, Spring House, Pennsylvania
| |
Collapse
|
32
|
Voigt B, Bhatia T, Hesselbarth J, Baumann M, Schmidt C, Ott M, Balbach J. The Prenucleation Equilibrium of the Parathyroid Hormone Determines the Critical Aggregation Concentration and Amyloid Fibril Nucleation. Chemphyschem 2023; 24:e202300439. [PMID: 37477386 DOI: 10.1002/cphc.202300439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/18/2023] [Indexed: 07/22/2023]
Abstract
Nucleation and growth of amyloid fibrils were found to only occur in supersaturated solutions above a critical concentration (ccrit ). The biophysical meaning of ccrit remained mostly obscure, since typical low values of ccrit in the sub-μM range hamper investigations of potential oligomeric states and their structure. Here, we investigate the parathyroid hormone PTH84 as an example of a functional amyloid fibril forming peptide with a comparably high ccrit of 67±21 μM. We describe a complex concentration dependent prenucleation ensemble of oligomers of different sizes and secondary structure compositions and highlight the occurrence of a trimer and tetramer at ccrit as possible precursors for primary fibril nucleation. Furthermore, the soluble state found in equilibrium with fibrils adopts to the prenucleation state present at ccrit . Our study sheds light onto early events of amyloid formation directly related to the critical concentration and underlines oligomer formation as a key feature of fibril nucleation. Our results contribute to a deeper understanding of the determinants of supersaturated peptide solutions. In the current study we present a biophysical approach to investigate ccrit of amyloid fibril formation of PTH84 in terms of secondary structure, cluster size and residue resolved intermolecular interactions during oligomer formation. Throughout the investigated range of concentrations (1 μM to 500 μM) we found different states of oligomerization with varying ability to contribute to primary fibril nucleation and with a concentration dependent equilibrium. In this context, we identified the previously described ccrit of PTH84 to mark a minimum concentration for the formation of homo-trimers/tetramers. These investigations allowed us to characterize molecular interactions of various oligomeric states that are further converted into elongation competent fibril nuclei during the lag phase of a functional amyloid forming peptide.
Collapse
Affiliation(s)
- Bruno Voigt
- Martin Luther University Halle-Wittenberg, Institute of Physics, Betty-Heimann-Straße 7, 06120, Halle, Germany
| | - Twinkle Bhatia
- Martin Luther University Halle-Wittenberg, Institute of Biochemistry and Biotechnology, Kurt-Mothes-Straße 3, 06120, Halle, Germany
| | - Julia Hesselbarth
- present address: Johannes Gutenberg University Mainz, Institute of Chemistry - Biochemistry, Biocenter II, Hanns-Dieter-Hüsch-Weg 17, 55128, Mainz, Germany
- Martin Luther University Halle-Wittenberg, Interdisciplinary Research Center HALOmem, Institute of Biochemistry and Biotechnology, Kurt-Mothes-Straße 3a, 06120, Halle, Germany
| | - Monika Baumann
- Martin Luther University Halle-Wittenberg, Institute of Physics, Betty-Heimann-Straße 7, 06120, Halle, Germany
| | - Carla Schmidt
- present address: Johannes Gutenberg University Mainz, Institute of Chemistry - Biochemistry, Biocenter II, Hanns-Dieter-Hüsch-Weg 17, 55128, Mainz, Germany
- Martin Luther University Halle-Wittenberg, Interdisciplinary Research Center HALOmem, Institute of Biochemistry and Biotechnology, Kurt-Mothes-Straße 3a, 06120, Halle, Germany
| | - Maria Ott
- Martin Luther University Halle-Wittenberg, Institute of Biochemistry and Biotechnology, Kurt-Mothes-Straße 3, 06120, Halle, Germany
| | - Jochen Balbach
- Martin Luther University Halle-Wittenberg, Institute of Physics, Betty-Heimann-Straße 7, 06120, Halle, Germany
| |
Collapse
|
33
|
Bhopatkar AA, Kayed R. Flanking regions, amyloid cores, and polymorphism: the potential interplay underlying structural diversity. J Biol Chem 2023; 299:105122. [PMID: 37536631 PMCID: PMC10482755 DOI: 10.1016/j.jbc.2023.105122] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/10/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023] Open
Abstract
The β-sheet-rich amyloid core is the defining feature of protein aggregates associated with neurodegenerative disorders. Recent investigations have revealed that there exist multiple examples of the same protein, with the same sequence, forming a variety of amyloid cores with distinct structural characteristics. These structural variants, termed as polymorphs, are hypothesized to influence the pathological profile and the progression of different neurodegenerative diseases, giving rise to unique phenotypic differences. Thus, identifying the origin and properties of these structural variants remain a focus of studies, as a preliminary step in the development of therapeutic strategies. Here, we review the potential role of the flanking regions of amyloid cores in inducing polymorphism. These regions, adjacent to the amyloid cores, show a preponderance for being structurally disordered, imbuing them with functional promiscuity. The dynamic nature of the flanking regions can then manifest in the form of conformational polymorphism of the aggregates. We take a closer look at the sequences flanking the amyloid cores, followed by a review of the polymorphic aggregates of the well-characterized proteins amyloid-β, α-synuclein, Tau, and TDP-43. We also consider different factors that can potentially influence aggregate structure and how these regions can be viewed as novel targets for therapeutic strategies by utilizing their unique structural properties.
Collapse
Affiliation(s)
- Anukool A Bhopatkar
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA.
| |
Collapse
|
34
|
Meyer N, Bentin J, Janot JM, Abrao-Nemeir I, Charles-Achille S, Pratlong M, Aquilina A, Trinquet E, Perrier V, Picaud F, Torrent J, Balme S. Ultrasensitive Detection of Aβ42 Seeds in Cerebrospinal Fluid with a Nanopipette-Based Real-Time Fast Amyloid Seeding and Translocation Assay. Anal Chem 2023; 95:12623-12630. [PMID: 37587130 DOI: 10.1021/acs.analchem.3c00017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
In this work, early-stage Aβ42 aggregates were detected using a real-time fast amyloid seeding and translocation (RT-FAST) assay. Specifically, Aβ42 monomers were incubated in buffer solution with and without preformed Aβ42 seeds in a quartz nanopipette coated with L-DOPA. Then, formed Aβ42 aggregates were analyzed on flyby resistive pulse sensing at various incubation time points. Aβ42 aggregates were detected only in the sample with Aβ42 seeds after 180 min of incubation, giving an on/off readout of the presence of preformed seeds. Moreover, this RT-FAST assay could detect preformed seeds spiked in 4% cerebrospinal fluid/buffer solution. However, in this condition, the time to detect the first aggregates was increased. Analysis of Cy3-labeled Aβ42 monomer adsorption on a quartz substrate after L-DOPA coating by confocal fluorescence spectroscopy and molecular dynamics simulation showed the huge influence of Aβ42 adsorption on the aggregation process.
Collapse
Affiliation(s)
- Nathan Meyer
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
- INM UM, CNRS, INSERM, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Jeremy Bentin
- Laboratoire de Nanomédecine, Imagerie et Thérapeutique, EA4662, Université Bourgogne-Franche-Comté (UFR Sciences et Techniques), Centre Hospitalier Universitaire de Besançon, 16 route de Gray, 25030 Besançon, France
| | - Jean-Marc Janot
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Imad Abrao-Nemeir
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Saly Charles-Achille
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Maud Pratlong
- PerkinElmer, Parc Marcel Boiteux, 30200 Codolet, France
| | | | - Eric Trinquet
- PerkinElmer, Parc Marcel Boiteux, 30200 Codolet, France
| | - Veronique Perrier
- INM UM, CNRS, INSERM, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Fabien Picaud
- Laboratoire de Nanomédecine, Imagerie et Thérapeutique, EA4662, Université Bourgogne-Franche-Comté (UFR Sciences et Techniques), Centre Hospitalier Universitaire de Besançon, 16 route de Gray, 25030 Besançon, France
| | - Joan Torrent
- INM UM, CNRS, INSERM, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| | - Sebastien Balme
- Institut Européen des Membranes, UMR5635 UM ENCSM CNRS, Place Eugène Bataillon, 34095 Montpellier cedex 5, France
| |
Collapse
|
35
|
Chandran A, Oliver HJ, Rochet JC. Role of NFE2L1 in the Regulation of Proteostasis: Implications for Aging and Neurodegenerative Diseases. BIOLOGY 2023; 12:1169. [PMID: 37759569 PMCID: PMC10525699 DOI: 10.3390/biology12091169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/29/2023]
Abstract
A hallmark of aging and neurodegenerative diseases is a disruption of proteome homeostasis ("proteostasis") that is caused to a considerable extent by a decrease in the efficiency of protein degradation systems. The ubiquitin proteasome system (UPS) is the major cellular pathway involved in the clearance of small, short-lived proteins, including amyloidogenic proteins that form aggregates in neurodegenerative diseases. Age-dependent decreases in proteasome subunit expression coupled with the inhibition of proteasome function by aggregated UPS substrates result in a feedforward loop that accelerates disease progression. Nuclear factor erythroid 2- like 1 (NFE2L1) is a transcription factor primarily responsible for the proteasome inhibitor-induced "bounce-back effect" regulating the expression of proteasome subunits. NFE2L1 is localized to the endoplasmic reticulum (ER), where it is rapidly degraded under basal conditions by the ER-associated degradation (ERAD) pathway. Under conditions leading to proteasome impairment, NFE2L1 is cleaved and transported to the nucleus, where it binds to antioxidant response elements (AREs) in the promoter region of proteasome subunit genes, thereby stimulating their transcription. In this review, we summarize the role of UPS impairment in aging and neurodegenerative disease etiology and consider the potential benefit of enhancing NFE2L1 function as a strategy to upregulate proteasome function and alleviate pathology in neurodegenerative diseases.
Collapse
Affiliation(s)
- Aswathy Chandran
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Haley Jane Oliver
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
36
|
Kaygisiz K, Rauch-Wirth L, Dutta A, Yu X, Nagata Y, Bereau T, Münch J, Synatschke CV, Weil T. Data-mining unveils structure-property-activity correlation of viral infectivity enhancing self-assembling peptides. Nat Commun 2023; 14:5121. [PMID: 37612273 PMCID: PMC10447463 DOI: 10.1038/s41467-023-40663-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 08/01/2023] [Indexed: 08/25/2023] Open
Abstract
Gene therapy via retroviral vectors holds great promise for treating a variety of serious diseases. It requires the use of additives to boost infectivity. Amyloid-like peptide nanofibers (PNFs) were shown to efficiently enhance retroviral gene transfer. However, the underlying mode of action of these peptides remains largely unknown. Data-mining is an efficient method to systematically study structure-function relationship and unveil patterns in a database. This data-mining study elucidates the multi-scale structure-property-activity relationship of transduction enhancing peptides for retroviral gene transfer. In contrast to previous reports, we find that not the amyloid fibrils themselves, but rather µm-sized β-sheet rich aggregates enhance infectivity. Specifically, microscopic aggregation of β-sheet rich amyloid structures with a hydrophobic surface pattern and positive surface charge are identified as key material properties. We validate the reliability of the amphiphilic sequence pattern and the general applicability of the key properties by rationally creating new active sequences and identifying short amyloidal peptides from various pathogenic and functional origin. Data-mining-even for small datasets-enables the development of new efficient retroviral transduction enhancers and provides important insights into the diverse bioactivity of the functional material class of amyloids.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Lena Rauch-Wirth
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstraße 1, 89081, Ulm, Germany
| | - Arghya Dutta
- Department Polymer Theory, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Xiaoqing Yu
- Department Molecular Spectroscopy, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Yuki Nagata
- Department Molecular Spectroscopy, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Tristan Bereau
- Department Polymer Theory, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- Institute for Theoretical Physics, Heidelberg University, Philosophenweg 19, 69120, Heidelberg, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstraße 1, 89081, Ulm, Germany
| | - Christopher V Synatschke
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany.
| | - Tanja Weil
- Department Synthesis of Macromolecules, Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany.
| |
Collapse
|
37
|
Vadukul D, Papp M, Thrush RJ, Wang J, Jin Y, Arosio P, Aprile FA. α-Synuclein Aggregation Is Triggered by Oligomeric Amyloid-β 42 via Heterogeneous Primary Nucleation. J Am Chem Soc 2023; 145:18276-18285. [PMID: 37556728 PMCID: PMC10450681 DOI: 10.1021/jacs.3c03212] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Indexed: 08/11/2023]
Abstract
An increasing number of cases where amyloids of different proteins are found in the same patient are being reported. This observation complicates diagnosis and clinical intervention. Amyloids of the amyloid-β peptide or the protein α-synuclein are traditionally considered hallmarks of Alzheimer's and Parkinson's diseases, respectively. However, the co-occurrence of amyloids of these proteins has also been reported in patients diagnosed with either disease. Here, we show that soluble species containing amyloid-β can induce the aggregation of α-synuclein. Fibrils formed under these conditions are solely composed of α-synuclein to which amyloid-β can be found associated but not as part of the core of the fibrils. Importantly, by global kinetic analysis, we found that the aggregation of α-synuclein under these conditions occurs via heterogeneous primary nucleation, triggered by soluble aggregates containing amyloid-β.
Collapse
Affiliation(s)
- Devkee
M. Vadukul
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| | - Marcell Papp
- Department
of Chemistry and Applied Biosciences, Institute
for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland
| | - Rebecca J. Thrush
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
- Institute
of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| | - Jielei Wang
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| | - Yiyun Jin
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| | - Paolo Arosio
- Department
of Chemistry and Applied Biosciences, Institute
for Chemical and Bioengineering, Swiss Federal Institute of Technology, 8093 Zurich, Switzerland
| | - Francesco A. Aprile
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
- Institute
of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.
| |
Collapse
|
38
|
Szała-Mendyk B, Phan TM, Mohanty P, Mittal J. Challenges in studying the liquid-to-solid phase transitions of proteins using computer simulations. Curr Opin Chem Biol 2023; 75:102333. [PMID: 37267850 PMCID: PMC10527940 DOI: 10.1016/j.cbpa.2023.102333] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/24/2023] [Accepted: 04/30/2023] [Indexed: 06/04/2023]
Abstract
"Membraneless organelles," also referred to as biomolecular condensates, perform a variety of cellular functions and their dysregulation is implicated in cancer and neurodegeneration. In the last two decades, liquid-liquid phase separation (LLPS) of intrinsically disordered and multidomain proteins has emerged as a plausible mechanism underlying the formation of various biomolecular condensates. Further, the occurrence of liquid-to-solid transitions within liquid-like condensates may give rise to amyloid structures, implying a biophysical link between phase separation and protein aggregation. Despite significant advances, uncovering the microscopic details of liquid-to-solid phase transitions using experiments remains a considerable challenge and presents an exciting opportunity for the development of computational models which provide valuable, complementary insights into the underlying phenomenon. In this review, we first highlight recent biophysical studies which provide new insights into the molecular mechanisms underlying liquid-to-solid (fibril) phase transitions of folded, disordered and multi-domain proteins. Next, we summarize the range of computational models used to study protein aggregation and phase separation. Finally, we discuss recent computational approaches which attempt to capture the underlying physics of liquid-to-solid transitions along with their merits and shortcomings.
Collapse
Affiliation(s)
- Beata Szała-Mendyk
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, TAMU 3127, College Station, 77843, Texas, United States.
| | - Tien Minh Phan
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, TAMU 3127, College Station, 77843, Texas, United States.
| | - Priyesh Mohanty
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, TAMU 3127, College Station, 77843, Texas, United States.
| | - Jeetain Mittal
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, TAMU 3127, College Station, 77843, Texas, United States; Department of Chemistry, Texas A&M University, TAMU 3255, College Station, 77843, Texas, United States; Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, TAMU 3255, College Station, 77843, Texas, United States.
| |
Collapse
|
39
|
Přáda Brichtová E, Krupová M, Bouř P, Lindo V, Gomes Dos Santos A, Jackson SE. Glucagon-like peptide 1 aggregates into low-molecular-weight oligomers off-pathway to fibrillation. Biophys J 2023; 122:2475-2488. [PMID: 37138517 PMCID: PMC10323027 DOI: 10.1016/j.bpj.2023.04.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/04/2023] [Accepted: 04/27/2023] [Indexed: 05/05/2023] Open
Abstract
The physical stability of peptide-based drugs is of great interest to the pharmaceutical industry. Glucagon-like peptide 1 (GLP-1) is a 31-amino acid peptide hormone, the analogs of which are frequently used in the treatment of type 2 diabetes. We investigated the physical stability of GLP-1 and its C-terminal amide derivative, GLP-1-Am, both of which aggregate into amyloid fibrils. While off-pathway oligomers have been proposed to explain the unusual aggregation kinetics observed previously for GLP-1 under specific conditions, these oligomers have not been studied in any detail. Such states are important as they may represent potential sources of cytotoxicity and immunogenicity. Here, we identified and isolated stable, low-molecular-weight oligomers of GLP-1 and GLP-1-Am, using size-exclusion chromatography. Under the conditions studied, isolated oligomers were shown to be resistant to fibrillation or dissociation. These oligomers contain between two and five polypeptide chains and they have a highly disordered structure as indicated by a variety of spectroscopic techniques. They are highly stable with respect to time, temperature, or agitation despite their noncovalent character, which was established using liquid chromatography-mass spectrometry and sodium dodecyl sulfate-polyacrylamide gel electrophoresis. These results provide evidence of stable, low-molecular-weight oligomers that are formed by an off-pathway mechanism which competes with amyloid fibril formation.
Collapse
Affiliation(s)
- Eva Přáda Brichtová
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Monika Krupová
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences, Prague 6, Czech Republic; Hylleraas Centre for Quantum Molecular Sciences, Department of Chemistry, UiT The Arctic University of Norway, Tromsø, Norway
| | - Petr Bouř
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences, Prague 6, Czech Republic
| | - Viv Lindo
- AstraZeneca, Cambridge, United Kingdom
| | | | - Sophie E Jackson
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
40
|
Limbocker R, Cremades N, Cascella R, Tessier PM, Vendruscolo M, Chiti F. Characterization of Pairs of Toxic and Nontoxic Misfolded Protein Oligomers Elucidates the Structural Determinants of Oligomer Toxicity in Protein Misfolding Diseases. Acc Chem Res 2023. [PMID: 37071750 DOI: 10.1021/acs.accounts.3c00045] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
ConspectusThe aberrant misfolding and aggregation of peptides and proteins into amyloid aggregates occurs in over 50 largely incurable protein misfolding diseases. These pathologies include Alzheimer's and Parkinson's diseases, which are global medical emergencies owing to their prevalence in increasingly aging populations worldwide. Although the presence of mature amyloid aggregates is a hallmark of such neurodegenerative diseases, misfolded protein oligomers are increasingly recognized as of central importance in the pathogenesis of many of these maladies. These oligomers are small, diffusible species that can form as intermediates in the amyloid fibril formation process or be released by mature fibrils after they are formed. They have been closely associated with the induction of neuronal dysfunction and cell death. It has proven rather challenging to study these oligomeric species because of their short lifetimes, low concentrations, extensive structural heterogeneity, and challenges associated with producing stable, homogeneous, and reproducible populations. Despite these difficulties, investigators have developed protocols to produce kinetically, chemically, or structurally stabilized homogeneous populations of protein misfolded oligomers from several amyloidogenic peptides and proteins at experimentally ameneable concentrations. Furthermore, procedures have been established to produce morphologically similar but structurally distinct oligomers from the same protein sequence that are either toxic or nontoxic to cells. These tools offer unique opportunities to identify and investigate the structural determinants of oligomer toxicity by a close comparative inspection of their structures and the mechanisms of action through which they cause cell dysfunction.This Account reviews multidisciplinary results, including from our own groups, obtained by combining chemistry, physics, biochemistry, cell biology, and animal models for pairs of toxic and nontoxic oligomers. We describe oligomers comprised of the amyloid-β peptide, which underlie Alzheimer's disease, and α-synuclein, which are associated with Parkinson's disease and other related neurodegenerative pathologies, collectively known as synucleinopathies. Furthermore, we also discuss oligomers formed by the 91-residue N-terminal domain of [NiFe]-hydrogenase maturation factor from E. coli, which we use as a model non-disease-related protein, and by an amyloid stretch of Sup35 prion protein from yeast. These oligomeric pairs have become highly useful experimental tools for studying the molecular determinants of toxicity characteristic of protein misfolding diseases. Key properties have been identified that differentiate toxic from nontoxic oligomers in their ability to induce cellular dysfunction. These characteristics include solvent-exposed hydrophobic regions, interactions with membranes, insertion into lipid bilayers, and disruption of plasma membrane integrity. By using these properties, it has been possible to rationalize in model systems the responses to pairs of toxic and nontoxic oligomers. Collectively, these studies provide guidance for the development of efficacious therapeutic strategies to target rationally the cytotoxicity of misfolded protein oligomers in neurodegenerative conditions.
Collapse
Affiliation(s)
- Ryan Limbocker
- Department of Chemistry and Life Science, United States Military Academy, West Point, New York 10996, United States
| | - Nunilo Cremades
- Institute for Biocomputation and Physics of Complex Systems (BIFI) and Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza 50009, Spain
| | - Roberta Cascella
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| | - Peter M Tessier
- Departments of Chemical Engineering, Pharmaceutical Sciences, and Biomedical Engineering, Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - Fabrizio Chiti
- Section of Biochemistry, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence 50134, Italy
| |
Collapse
|
41
|
Yoon J, Lee M, Park Y, Lee K, Shin S. In silico investigation of the structural stability as the origin of the pathogenicity of α-synuclein protofibrils. J Biomol Struct Dyn 2023; 41:14103-14115. [PMID: 37036430 DOI: 10.1080/07391102.2023.2199077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/31/2023] [Indexed: 04/11/2023]
Abstract
α-Synuclein is a presynaptic neuronal protein. The fibril form of α-synuclein is a major constituent of the intraneuronal inclusion called Lewy body, a characteristic hallmark of Parkinson's disease. Recent ssNMR and cryo-EM experiments of wild-type α-synuclein fibrils have shown polymorphism and observed two major polymorphs, rod and twister. To associate the cytotoxicity of α-synuclein fibrils with their structural features, it is essential to understand the origins of their structural stability. In this study, we performed molecular dynamics simulations of the two major polymorphs of wild-type α-synuclein fibrils. The predominance of specific fibril polymorphs was rationalized in terms of relative structural stability in aqueous environments, which was attributed to the cooperative contributions of various stabilizing features. The results of the simulations indicated that highly stable structures in aqueous environments could be maintained by the cooperation of compact sidechain packing in the hydrophobic core, backbone geometry of the maximal β-sheet content wrapping the hydrophobic core, and solvent-exposed sidechains with large fluctuations maximizing the solvation entropy. The paired structure of the two protofilaments provides additional stability, especially at the interface region, by forming steric zipper interactions and hiding the hydrophobic residues from exposure to water. The sidechain interaction analyses and pulling simulations showed that the rod polymorph has stronger sidechain interactions and exhibits higher dissociation energy than the twister polymorph. It is expected that our study will provide a basis for understanding the pathogenic behaviors of diverse amyloid strains in terms of their structural properties.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jeseong Yoon
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - MinJun Lee
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Yunsu Park
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Kyunghee Lee
- Department of Chemistry, Sejong University, Seoul, Republic of Korea
| | - Seokmin Shin
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
42
|
Morfino P, Aimo A, Vergaro G, Sanguinetti C, Castiglione V, Franzini M, Perrone MA, Emdin M. Transthyretin Stabilizers and Seeding Inhibitors as Therapies for Amyloid Transthyretin Cardiomyopathy. Pharmaceutics 2023; 15:pharmaceutics15041129. [PMID: 37111614 PMCID: PMC10143494 DOI: 10.3390/pharmaceutics15041129] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/15/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Transthyretin (TTR) amyloid cardiomyopathy (ATTR-CM) is a progressive and increasingly recognized cause of heart failure which is associated with high mortality and morbidity. ATTR-CM is characterized by the misfolding of TTR monomers and their deposition within the myocardium as amyloid fibrils. The standard of care for ATTR-CM consists of TTR-stabilizing ligands, such as tafamidis, which aim at maintaining the native structure of TTR tetramers, thus preventing amyloid aggregation. However, their efficacy in advanced-staged disease and after long-term treatment is still a source of concern, suggesting the existence of other pathogenetic factors. Indeed, pre-formed fibrils present in the tissue can further accelerate amyloid aggregation in a self-propagating process known as “amyloid seeding”. The inhibition of amyloidogenesis through TTR stabilizers combined with anti-seeding peptides may represent a novel strategy with additional benefits over current therapies. Finally, the role of stabilizing ligands needs to be reassessed in view of the promising results derived from trials which have evaluated alternative strategies, such as TTR silencers and immunological amyloid disruptors.
Collapse
Affiliation(s)
- Paolo Morfino
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Alberto Aimo
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| | - Giuseppe Vergaro
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| | - Chiara Sanguinetti
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Vincenzo Castiglione
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| | - Maria Franzini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Michele Emdin
- Interdisciplinary Center for Health Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, 56124 Pisa, Italy
| |
Collapse
|
43
|
Tammara V, Das A. Governing dynamics and preferential binding of the AXH domain influence the aggregation pathway of Ataxin-1. Proteins 2023; 91:380-394. [PMID: 36208132 DOI: 10.1002/prot.26436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 08/30/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
The present state of understanding the mechanism of Spinocerebellar Ataxia-1, a fatal neurodegenerative disease linked to the protein Ataxin-1 (ATXN1), is baffled by a set of self-contradictory, and hence, inconclusive observations. This fallacy poses a bottleneck to the effective designing of curable drugs as the field is currently missing the specific druggable site. To understand the fundamentals of pathogenesis, we tried to decipher the intricacies of the extremely complicated landscape by targeting the relevant species that supposedly dictate the structure-function paradigm. The atomic-level description and characterization of the dynamism of the systems reveal the existence of structural polymorphism in all the leading stakeholders of the overall system. The very existence of conformational heterogeneity in every species creates numerous possible combinations of favorable interactions because of the variability in segmental cross-talks and hence claims its role in the choice of routes between functional activity and dysfunctional disease-causing aggregation. Despite this emergent configurational diversity, there is a common mode of operative intermolecular forces that dictates the extent of stability of all the multimeric complexes due to the localized population of a specific type of residue. The present research proposes a dynamic switch mechanism between aggregability and functional activity, based on the logical interpretation of the estimated variables, which is practically dictated by the effective concentration of the interacting species involved in the cell.
Collapse
Affiliation(s)
- Vaishnavi Tammara
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Atanu Das
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
44
|
Habashi M, Chauhan PS, Vutla S, Senapati S, Diachkov M, El-Husseini A, Guérin B, Lubell WD, Rahimipour S. Aza-Residue Modulation of Cyclic d,l-α-Peptide Nanotube Assembly with Enhanced Anti-Amyloidogenic Activity. J Med Chem 2023; 66:3058-3072. [PMID: 36763536 DOI: 10.1021/acs.jmedchem.2c02049] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Transient soluble oligomers of amyloid-β (Aβ) are considered among the most toxic species in Alzheimer's disease (AD). Soluble Aβ oligomers accumulate early prior to insoluble plaque formation and cognitive impairment. The cyclic d,l-α-peptide CP-2 (1) self-assembles into nanotubes and demonstrates promising anti-amyloidogenic activity likely by a mechanism involving engagement of soluble oligomers. Systematic replacement of the residues in peptide 1 with aza-amino acid counterparts was performed to explore the effects of hydrogen bonding on propensity to mitigate Aβ aggregation and toxicity. Certain azapeptides exhibited improved ability to engage, alter the secondary structure, and inhibit aggregation of Aβ. Moreover, certain azapeptides disassembled preformed Aβ fibrils and protected cells from Aβ-mediated toxicity. Substitution of the l-norleucine3 and d-serine6 residues in peptide 1 with aza-norleucine and aza-homoserine provided, respectively, nontoxic [azaNle3]-1 (4) and [azaHse6]-1 (7), that significantly abated symptoms in a transgenic Caenorhabditis elegans AD model by decreasing Aβ oligomer levels.
Collapse
Affiliation(s)
- Maram Habashi
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Pradeep S Chauhan
- Département de Chimie, Université de Montréal, Complexe des Sciences, B-3015 1375 Avenue Thérèse-Lavoie-Roux, Montréal, Québec H2V 0B3, Canada
| | - Suresh Vutla
- Département de Chimie, Université de Montréal, Complexe des Sciences, B-3015 1375 Avenue Thérèse-Lavoie-Roux, Montréal, Québec H2V 0B3, Canada
| | - Sudipta Senapati
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Mykhailo Diachkov
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Ali El-Husseini
- Département de Chimie, Université de Montréal, Complexe des Sciences, B-3015 1375 Avenue Thérèse-Lavoie-Roux, Montréal, Québec H2V 0B3, Canada
| | - Brigitte Guérin
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke 3001, 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
- Sherbrooke Molecular Imaging Center (CIMS), Research centre of the CHUS (CRCHUS) 3001, 12e Avenue Nord, Sherbrooke, Quebec J1H 5N4, Canada
| | - William D Lubell
- Département de Chimie, Université de Montréal, Complexe des Sciences, B-3015 1375 Avenue Thérèse-Lavoie-Roux, Montréal, Québec H2V 0B3, Canada
| | - Shai Rahimipour
- Department of Chemistry, Bar-Ilan University, Ramat-Gan 5290002, Israel
| |
Collapse
|
45
|
Yi Y, Lim MH. Current understanding of metal-dependent amyloid-β aggregation and toxicity. RSC Chem Biol 2023; 4:121-131. [PMID: 36794021 PMCID: PMC9906324 DOI: 10.1039/d2cb00208f] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022] Open
Abstract
The discovery of effective therapeutics targeting amyloid-β (Aβ) aggregates for Alzheimer's disease (AD) has been very challenging, which suggests its complicated etiology associated with multiple pathogenic elements. In AD-affected brains, highly concentrated metals, such as copper and zinc, are found in senile plaques mainly composed of Aβ aggregates. These metal ions are coordinated to Aβ and affect its aggregation and toxicity profiles. In this review, we illustrate the current view on molecular insights into the assembly of Aβ peptides in the absence and presence of metal ions as well as the effect of metal ions on their toxicity.
Collapse
Affiliation(s)
- Yelim Yi
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST) Daejeon 34141 Republic of Korea
| |
Collapse
|
46
|
Pauly T, Zhang T, Sternke-Hoffmann R, Nagel-Steger L, Willbold D. Differentiation of subnucleus-sized oligomers and nucleation-competent assemblies of the Aβ peptide. Biophys J 2023; 122:269-278. [PMID: 36529991 PMCID: PMC9892607 DOI: 10.1016/j.bpj.2022.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/16/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
A significant feature of Alzheimer's disease is the formation of amyloid deposits in the brain consisting mainly of misfolded derivatives of proteolytic cleavage products of the amyloid precursor protein amyloid-β (Aβ) peptide. While high-resolution structures already exist for both the monomer and the amyloid fibril of the Aβ peptide, the mechanism of amyloid formation itself still defies precise characterization. In this study, low and high molecular weight oligomers (LMWOs and HMWOs) were identified by sedimentation velocity analysis, and for the first time, the temporal evolution of oligomer size distributions was correlated with the kinetics of amyloid formation as determined by thioflavin T-binding studies. LMWOs of subnucleus size contain fewer than seven monomer units and exist alongside a heterogeneous group of HMWOs with 20-160 monomer units that represent potential centers of nucleus formation due to high local monomer concentrations. These HMWOs already have slightly increased β-strand content and appear structurally similar regardless of size, as shown by examination with a range of fluorescent dyes. Once fibril nuclei are formed, the monomer concentration begins to decrease, followed by a decrease in oligomer concentration, starting with LMWOs, which are the least stable species. The observed behavior classifies the two LMWOs as off pathway. In contrast, we consider HMWOs to be on-pathway, prefibrillar intermediates, representing structures in which nucleated conformational conversion is facilitated by high local concentrations. Aβ40 and Aβ42 M35ox take much longer to form nuclei and enter the growth phase than Aβ42 under identical reaction conditions, presumably because both the size and the concentration of HMWOs formed are much smaller.
Collapse
Affiliation(s)
- Thomas Pauly
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Research Center Jülich, Jülich, Germany
| | - Tao Zhang
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Research Center Jülich, Jülich, Germany
| | | | - Luitgard Nagel-Steger
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Research Center Jülich, Jülich, Germany.
| | - Dieter Willbold
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany; Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Research Center Jülich, Jülich, Germany
| |
Collapse
|
47
|
Liquid-liquid Phase Separation of α-Synuclein: A New Mechanistic Insight for α-Synuclein Aggregation Associated with Parkinson's Disease Pathogenesis. J Mol Biol 2023; 435:167713. [PMID: 35787838 DOI: 10.1016/j.jmb.2022.167713] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023]
Abstract
Aberrant aggregation of the misfolded presynaptic protein, α-Synuclein (α-Syn) into Lewy body (LB) and Lewy neuritis (LN) is a major pathological hallmark of Parkinson's disease (PD) and other synucleinopathies. Numerous studies have suggested that prefibrillar and fibrillar species of the misfolded α-Syn aggregates are responsible for cell death in PD pathogenesis. However, the precise molecular events during α-Syn aggregation, especially in the early stages, remain elusive. Emerging evidence has demonstrated that liquid-liquid phase separation (LLPS) of α-Syn occurs in the nucleation step of α-Syn aggregation, which offers an alternate non-canonical aggregation pathway in the crowded microenvironment. The liquid-like α-Syn droplets gradually undergo an irreversible liquid-to-solid phase transition into amyloid-like hydrogel entrapping oligomers and fibrils. This new mechanism of α-Syn LLPS and gel formation might represent the molecular basis of cellular toxicity associated with PD. This review aims to demonstrate the recent development of α-Syn LLPS, the underlying mechanism along with the microscopic events of aberrant phase transition. This review further discusses how several intrinsic and extrinsic factors regulate the thermodynamics and kinetics of α-Syn LLPS and co-LLPS with other proteins, which might explain the pathophysiology of α-Syn in various neurodegenerative diseases.
Collapse
|
48
|
Kwak J, Woo J, Park S, Lim MH. Rational design of photoactivatable metal complexes to target and modulate amyloid-β peptides. J Inorg Biochem 2023; 238:112053. [PMID: 36347209 DOI: 10.1016/j.jinorgbio.2022.112053] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/25/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022]
Abstract
The accumulation of amyloid-β (Aβ) aggregates is found in the brains of Alzheimer's disease patients. Thus, numerous efforts have been made to develop chemical reagents capable of targeting Aβ peptides and controlling their aggregation. In particular, tunable coordination and photophysical properties of transition metal complexes, with variable oxidation and spin states on the metal centers, can be utilized to probe Aβ aggregates and alter their aggregation profiles. In this review, we illustrate some rational strategies for designing photoactivatable metal complexes as chemical sensors for Aβ peptides or modulators against their aggregation pathways, with some examples.
Collapse
Affiliation(s)
- Jimin Kwak
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Junhyeok Woo
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seongmin Park
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Mi Hee Lim
- Department of Chemistry, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
49
|
Grønnemose AL, Østerlund EC, Otzen DE, Jørgensen TJD. EGCG has Dual and Opposing Effects on the N-terminal Region of Self-associating α-synuclein Oligomers. J Mol Biol 2022; 434:167855. [PMID: 36240861 DOI: 10.1016/j.jmb.2022.167855] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/11/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Oligomers of the protein α-synuclein (α-syn) are thought to be a major toxic species in Parkinson's disease, particularly through their ability to permeabilize cell membranes. The green tea polyphenol epigallocatechin gallate (EGCG) has been found to reduce this ability. We have analyzed α-syn oligomer dynamics and interconversion by H/D exchange monitored by mass spectrometry (HDX-MS). Our results show that the two oligomers OI and OII co-exist in equilibrium; OI is a multimer of OII and its dissociation can be followed by HDX-MS by virtue of the correlated exchange of the N-terminal region. Urea destabilizes the α-syn oligomers, dissociating OI to OII and monomers. Oligomers exposed to EGCG undergo Met oxidation. Intriguingly, EGCG induces an oxidation-dependent effect on the structure of the N-terminal region. For the non-oxidized N-terminal region, EGCG increases the stability of the folded structure as measured by a higher level of protection against H/D exchange. In contrast, protection is clearly abrogated in the Met oxidized N-terminal region. Having a non-oxidized and disordered N-terminal region is known to be essential for efficient membrane binding. Therefore, our results suggest that the combined effect of a structural stabilization of the non-oxidized N-terminal region and the presence of a disordered oxidized N-terminal region renders the oligomers less cytotoxic by decreasing the ability of the N-terminal region to bind to cell membranes and facilitate their permeabilization.
Collapse
Affiliation(s)
- Anne Louise Grønnemose
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark; Department of Biochemistry and Molecular Biology (BMB), University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Eva Christina Østerlund
- Department of Biochemistry and Molecular Biology (BMB), University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Daniel Erik Otzen
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark.
| | - Thomas J D Jørgensen
- Department of Biochemistry and Molecular Biology (BMB), University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| |
Collapse
|
50
|
Wu K, Sun W, Li D, Diao J, Xiu P. Inhibition of Amyloid Nucleation by Steric Hindrance. J Phys Chem B 2022; 126:10045-10054. [PMID: 36417323 DOI: 10.1021/acs.jpcb.2c06330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Despite recent experiments and simulations suggesting that small-molecule inhibitors and some post-translational modifications (e.g., glycosylation and ubiquitination) can suppress the pathogenic aggregation of proteins due to steric hindrance, the effect of steric hindrance on amyloid formation has not been systematically studied. Based on Monte Carlo simulations using a coarse-grained model for amyloidogenic proteins and a hard sphere acting as steric hindrance, we investigated how steric hindrance on proteins could affect amyloid formation, particularly two steps of primary nucleation, namely, oligomerization and conformational conversion into a β-sheet-enriched nucleus. We found that steric spheres played an inhibitory role in oligomerization with the effect proportional to the sphere radius RS, which we attributed to the decline in the nonspecific attractions between proteins. During the second step, small steric spheres facilitated the conformational conversion of proteins while large ones suppressed the conversion. The overall steric effect on amyloid nucleation was inhibitory regardless of RS. As RS increased, oligomeric assemblies changed from amorphous into sheet-like, structurally ordered species, reminiscent of the structure of amyloid fibrils. The oligomers with large RS were off-pathway with their ordered structures induced by the competition between steric hindrance and nonspecific attractions of soluble proteins. Interestingly, the equimolar mixture of proteins with and without steric hindrance amplified the sterically inhibitory effect by increasing the energy barrier of protein's conformational conversion. The physical mechanisms and biological implications of the above results are discussed. Our findings improve the current understanding of how nature regulates protein aggregation and amyloid formation by steric hindrance.
Collapse
Affiliation(s)
- Kai Wu
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, People's Republic of China.,School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.,Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China.,Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, United States of America
| | - Wuxuepeng Sun
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Dechang Li
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, United States of America
| | - Peng Xiu
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, People's Republic of China
| |
Collapse
|