1
|
Brennan GS, Goriely A. A network aggregation model for amyloid- β dynamics and treatment of Alzheimer's diseases at the brain scale. J Math Biol 2025; 90:22. [PMID: 39891738 PMCID: PMC11787187 DOI: 10.1007/s00285-024-02179-5] [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: 07/01/2024] [Revised: 10/22/2024] [Accepted: 12/22/2024] [Indexed: 02/03/2025]
Abstract
Neurodegenerative diseases are associated with the assembly of specific proteins into oligomers and fibrillar aggregates. At the brain scale, these protein assemblies can diffuse through the brain and seed other regions, creating an autocatalytic protein progression. The growth and transport of these assemblies depend on various mechanisms that can be targeted therapeutically. Here, we use spatially-extended nucleation-aggregation-fragmentation models for the dynamics of prion-like neurodegenerative protein-spreading in the brain to study the effect of different drugs on whole-brain Alzheimer's disease progression.
Collapse
Affiliation(s)
- Georgia S Brennan
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Rd, Oxford, OX2 6GG, UK
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Rd, Oxford, OX2 6GG, UK.
| |
Collapse
|
2
|
Silva A, Duarte-Silva S, Martins PM, Rodrigues B, Serrenho D, Vilasboas-Campos D, Teixeira-Castro A, Vieyto-Nuñez J, Mieres-Perez J, Figueiredo F, Fraga J, Noble J, Lantz C, Sepanj N, Monteiro-Fernandes D, Guerreiro S, Neves-Carvalho A, Pereira-Sousa J, Klärner FG, Schrader T, Loo JA, Pastore A, Sanchez-Garcia E, Bitan G, Carvalho AL, Maciel P, Macedo-Ribeiro S. Allosteric Modulation of Pathological Ataxin-3 Aggregation: A Path to Spinocerebellar Ataxia Type-3 Therapies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.633970. [PMID: 39896516 PMCID: PMC11785186 DOI: 10.1101/2025.01.22.633970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Spinocerebellar ataxia type 3 (SCA3) is a rare inherited neurodegenerative disease caused by the expansion of a polyglutamine repeat in the protease ataxin-3 (Atx3). Despite extensive knowledge of the downstream pathophysiology, no disease-modifying therapies are currently available to halt disease progression. The accumulation of protein inclusions enriched in the polyQ-expanded Atx3 in neurons suggests that inhibiting its self-assembly may yield targeted therapeutic approaches. Here it is shown that a supramolecular tweezer, CLR01, binds to a lysine residue on a positively charged surface patch of the Atx3 catalytic Josephin domain. At this site, the binding of CLR01 decreases the conformational fluctuations of the distal flexible hairpin. This results in reduced exposure of the nearby aggregation-prone region, which overlaps with the substrate ubiquitin binding site and primes Atx3 self-assembly, ultimately delaying Atx3 amyloid fibril formation and reducing the secondary nucleation rate, a process linked to fibril proliferation and toxicity. These effects translate into the reversal of synapse loss in a SCA3 cultured cortical neuron model, an improved locomotor function in a C. elegans SCA3 model, and a delay in disease onset, accompanied by reduced severity of motor symptoms in a SCA3 mouse model. This study provides critical insights into Atx3 self-assembly, revealing a novel allosteric site for designing CLR01-inspired therapies targeting pathological aggregation pathways while sparing essential functional sites. These findings emphasize that targeting allosteric sites in amyloid-forming proteins may offer unique opportunities to develop safe therapeutic strategies for various protein misfolding disorders.
Collapse
Affiliation(s)
- Alexandra Silva
- i3S -Institute for Research and Innovation in Health, Porto University, Porto, Portugal
- Institute for Molecular and Cellular Biology (IBMC), Porto University, Porto, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Pedro M Martins
- i3S -Institute for Research and Innovation in Health, Porto University, Porto, Portugal
- Institute for Molecular and Cellular Biology (IBMC), Porto University, Porto, Portugal
| | - Beatriz Rodrigues
- Center for Neuroscience and Cell Biology (CNC-UC) & Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Débora Serrenho
- Center for Neuroscience and Cell Biology (CNC-UC) & Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Daniela Vilasboas-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Julio Vieyto-Nuñez
- Department of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
| | - Joel Mieres-Perez
- Department of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
| | - Francisco Figueiredo
- i3S -Institute for Research and Innovation in Health, Porto University, Porto, Portugal
- Institute for Molecular and Cellular Biology (IBMC), Porto University, Porto, Portugal
| | - Joana Fraga
- i3S -Institute for Research and Innovation in Health, Porto University, Porto, Portugal
- Institute for Molecular and Cellular Biology (IBMC), Porto University, Porto, Portugal
| | - James Noble
- King's College London, London, United Kingdom
| | - Carter Lantz
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | - Niki Sepanj
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniela Monteiro-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Sara Guerreiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Andreia Neves-Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Joana Pereira-Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | | | - Thomas Schrader
- Faculty of Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Joseph A Loo
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA, USA
| | | | - Elsa Sanchez-Garcia
- Department of Biochemical and Chemical Engineering, TU Dortmund University, Dortmund, Germany
| | - Gal Bitan
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Brain Research Institute and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ana Luísa Carvalho
- Center for Neuroscience and Cell Biology (CNC-UC) & Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B's, PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Sandra Macedo-Ribeiro
- i3S -Institute for Research and Innovation in Health, Porto University, Porto, Portugal
- Institute for Molecular and Cellular Biology (IBMC), Porto University, Porto, Portugal
| |
Collapse
|
3
|
Kotarba S, Kozłowska M, Scios M, Saramowicz K, Barczuk J, Granek Z, Siwecka N, Wiese W, Golberg M, Galita G, Sychowski G, Majsterek I, Rozpędek-Kamińska W. Potential Mechanisms of Tunneling Nanotube Formation and Their Role in Pathology Spread in Alzheimer's Disease and Other Proteinopathies. Int J Mol Sci 2024; 25:10797. [PMID: 39409126 PMCID: PMC11477428 DOI: 10.3390/ijms251910797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia worldwide. The etiopathogenesis of this disease remains unknown. Currently, several hypotheses attempt to explain its cause, with the most well-studied being the cholinergic, beta-amyloid (Aβ), and Tau hypotheses. Lately, there has been increasing interest in the role of immunological factors and other proteins such as alpha-synuclein (α-syn) and transactive response DNA-binding protein of 43 kDa (TDP-43). Recent studies emphasize the role of tunneling nanotubes (TNTs) in the spread of pathological proteins within the brains of AD patients. TNTs are small membrane protrusions composed of F-actin that connect non-adjacent cells. Conditions such as pathogen infections, oxidative stress, inflammation, and misfolded protein accumulation lead to the formation of TNTs. These structures have been shown to transport pathological proteins such as Aβ, Tau, α-syn, and TDP-43 between central nervous system (CNS) cells, as confirmed by in vitro studies. Besides their role in spreading pathology, TNTs may also have protective functions. Neurons burdened with α-syn can transfer protein aggregates to glial cells and receive healthy mitochondria, thereby reducing cellular stress associated with α-syn accumulation. Current AD treatments focus on alleviating symptoms, and clinical trials with Aβ-lowering drugs have proven ineffective. Therefore, intensifying research on TNTs could bring scientists closer to a better understanding of AD and the development of effective therapies.
Collapse
Affiliation(s)
- Szymon Kotarba
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Marta Kozłowska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Małgorzata Scios
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Kamil Saramowicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Julia Barczuk
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Zuzanna Granek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Michał Golberg
- Department of Histology and Embryology, Medical University of Lodz, 90-419 Lodz, Poland;
| | - Grzegorz Galita
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Grzegorz Sychowski
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (S.K.); (M.K.); (M.S.); (K.S.); (J.B.); (Z.G.); (N.S.); (W.W.); (G.G.); (G.S.); (I.M.)
| |
Collapse
|
4
|
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
|
5
|
Lim S, Cho Y, Kang JH, Hwang M, Park Y, Kwak SK, Jung SH, Jung JH. Metallosupramolecular Multiblock Copolymers of Lanthanide Complexes by Seeded Living Polymerization. J Am Chem Soc 2024; 146:18484-18497. [PMID: 38888168 DOI: 10.1021/jacs.4c03983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Supramolecular block copolymers, derived via seeded living polymerization, are increasingly recognized for their rich structural and functional diversity, marking them as cutting-edge materials. The use of metal complexes in supramolecular block copolymerization not only offers a broad range of block copolymers through the structural similarity in the coordination geometry of the central metal ion but also controls spectroscopic properties, such as emission wavelength, emission strength, and fluorescence lifetime. However, the exploration of metallosupramolecular multiblock copolymerization based on metal complexes remains quite limited. In this work, we present a pioneering synthesis of metallosupramolecular multiblock copolymers utilizing Eu3+ and Tb3+ complexes as building blocks. This is achieved through the strategic manipulation of nonequilibrium self-assemblies via a living supramolecular polymerization approach. Our comprehensive exploration of both thermodynamically and kinetically regulated metallosupramolecular polymerizations, centered around Eu3+ and Tb3+ complexes with bisterpyridine-modified ligands containing R-alanine units and a long alkyl group, has highlighted intriguing behaviors. The monomeric [R-L1Eu(NO3)3] complex generates a spherical structure as the kinetic product. In contrast, the monomeric [R-L1Eu2(NO3)6] complex generates fiber aggregates as a thermodynamic product through intermolecular interactions such as π-π stacking, hydrophobic interaction, and H-bonds. Utilizing the Eu3+ complex, we successfully conducted seed-induced living polymerization of the monomeric building unit under kinetically regulated conditions. This yielded a metallosupramolecular polymer of precisely controlled length with minimal polydispersity. Moreover, by copolymerizing the kinetically confined Tb3+ complex state ("A" species) with a seed derived from the Eu3+ complex ("B" species), we were able to fabricate metallosupramolecular tri- and pentablock copolymers with A-B-A, and B-A-B-A-B types, respectively, through a seed-end chain-growth mechanism.
Collapse
Affiliation(s)
- Seola Lim
- Department of Chemistry, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Yumi Cho
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Ju Hwan Kang
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Minkyeong Hwang
- Department of Chemistry, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Yumi Park
- Department of Chemistry, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Sang Kyu Kwak
- Department of Chemical and Biological Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Sung Ho Jung
- Department of Chemistry, Gyeongsang National University, Jinju 52828, Republic of Korea
- Research Institute of Advanced Chemistry, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jong Hwa Jung
- Department of Chemistry, Gyeongsang National University, Jinju 52828, Republic of Korea
- Research Institute of Advanced Chemistry, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Frankel R, Sparr E, Linse S. Retardation of Aβ42 fibril formation by apolipoprotein A-I and recombinant HDL particles. J Biol Chem 2023; 299:105273. [PMID: 37739034 PMCID: PMC10616404 DOI: 10.1016/j.jbc.2023.105273] [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: 03/03/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
The double nucleation mechanism of amyloid β (Aβ) peptide aggregation is retained from buffer to cerebrospinal fluid (CSF) but with reduced rate of all microscopic processes. Here, we used a bottom-up approach to identify retarding factors in CSF. We investigated the Aβ42 fibril formation as a function of time in the absence and presence of apolipoprotein A-I (ApoA-I), recombinant high-density lipoprotein (rHDL) particles, or lipid vesicles. A retardation was observed in the presence of ApoA-I or rHDL particles, most pronounced with ApoA-I, but not with lipid vesicles. Global kinetic analysis implies that rHDL interferes with secondary nucleation. The effect of ApoA-I could best be described as an interference with secondary and to a smaller extent primary nucleation. Using surface plasmon resonance and microfluidics diffusional sizing analyses, we find that both rHDL and ApoA-I interact with Aβ42 fibrils but not Aβ42 monomer, thus the effect on kinetics seems to involve interference with the catalytic surface for secondary nucleation. The Aβ42 fibrils were imaged using cryogenic-electron microscopy and found to be longer when formed in the presence of ApoA-I or rHDL, compared to formation in buffer. A retarding effect, as observed in CSF, could be replicated using a simpler system, from key components present in CSF but purified from a CSF-free host. However, the effect of CSF is stronger implying the presence of additional retarding factors.
Collapse
Affiliation(s)
- Rebecca Frankel
- Biochemistry and Structural Biology, Lund University, Lund, Sweden; Division of Physical Chemistry, Lund University, Lund, Sweden
| | - Emma Sparr
- Division of Physical Chemistry, Lund University, Lund, Sweden
| | - Sara Linse
- Biochemistry and Structural Biology, Lund University, Lund, Sweden.
| |
Collapse
|
9
|
Österlund N, Frankel R, Carlsson A, Thacker D, Karlsson M, Matus V, Gräslund A, Emanuelsson C, Linse S. The C-terminal domain of the antiamyloid chaperone DNAJB6 binds to amyloid-β peptide fibrils and inhibits secondary nucleation. J Biol Chem 2023; 299:105317. [PMID: 37797698 PMCID: PMC10641233 DOI: 10.1016/j.jbc.2023.105317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 10/07/2023] Open
Abstract
The DNAJB6 chaperone inhibits fibril formation of aggregation-prone client peptides through interaction with aggregated and oligomeric forms of the amyloid peptides. Here, we studied the role of its C-terminal domain (CTD) using constructs comprising either the entire CTD or the first two or all four of the CTD β-strands grafted onto a scaffold protein. Each construct was expressed as WT and as a variant with alanines replacing five highly conserved and functionally important serine and threonine residues in the first β-strand. We investigated the stability, oligomerization, antiamyloid activity, and affinity for amyloid-β (Aβ42) species using optical spectroscopy, native mass spectrometry, chemical crosslinking, and surface plasmon resonance technology. While DNAJB6 forms large and polydisperse oligomers, CTD was found to form only monomers, dimers, and tetramers of low affinity. Kinetic analyses showed a shift in inhibition mechanism. Whereas full-length DNAJB6 activity is dependent on the serine and threonine residues and efficiently inhibits primary and secondary nucleation, all CTD constructs inhibit secondary nucleation only, independently of the serine and threonine residues, although their dimerization and thermal stabilities are reduced by alanine substitution. While the full-length DNAJB6 inhibition of primary nucleation is related to its propensity to form coaggregates with Aβ, the CTD constructs instead bind to Aβ42 fibrils, which affects the nucleation events at the fibril surface. The retardation of secondary nucleation by DNAJB6 can thus be ascribed to the first two β-strands of its CTD, whereas the inhibition of primary nucleation is dependent on the entire protein or regions outside the CTD.
Collapse
Affiliation(s)
- Nicklas Österlund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
| | - Rebecca Frankel
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Andreas Carlsson
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Dev Thacker
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Maja Karlsson
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Vanessa Matus
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Astrid Gräslund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Cecilia Emanuelsson
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden
| | - Sara Linse
- Division of Biochemistry and Structural Biology, Department of Chemistry, Lund University, Lund, Sweden.
| |
Collapse
|
10
|
De Sio S, Waegele J, Bhatia T, Voigt B, Lilie H, Ott M. Inherent Adaptivity of Alzheimer Peptides to Crowded Environments. Macromol Biosci 2023; 23:e2200527. [PMID: 37066978 DOI: 10.1002/mabi.202200527] [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: 11/30/2022] [Revised: 03/30/2023] [Indexed: 04/18/2023]
Abstract
Amyloid β (Aβ) is the major constituent in senile plaques of Alzheimer's disease in which peptides initially undergo structural conversions to form elongated fibrils. The impact of crowding on the fibrillation pathways of Aβ40 and Aβ42 , the most common peptide isoforms are studied. PEG and Ficoll are used as model crowders to mimic a macromolecular enriched surrounding. The fibrillar growth is monitored with the help of ThT-fluorescence assays in order to extract two rates describing primary and secondary processes of nucleation and growth. Techniques as fluorescence correlation spectroscopy and analytical ultracentrifugation are used to discuss oligomeric states; fibril morphologies are investigated using negative-staining transmission electron microscopy. While excluded volume effects imposed by macromolecular crowding are expected to always increase rates of intermolecular interactions and structural conversion, a vast variety of effects are found depending on the peptide, the crowder, or ionic strength of the solution. While investigations of the obtained rates with respect to a reactant-occluded model are capable to display specific surface interactions with the crowder, the employment of crystallization-like models reveal the crowder-induced entropic gain withΔ Δ G fib crow = - 116 ± 21 k $\Delta \Delta G_{\text{fib}}^{\text{crow}}=-116\pm 21\; k$ J mol-1 per volume fraction of the crowder.
Collapse
Affiliation(s)
- Silvia De Sio
- Department of Biotechnology and Biochemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, Halle, 06120, Saxony-Anhalt, Germany
| | - Jana Waegele
- Department of Biotechnology and Biochemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, Halle, 06120, Saxony-Anhalt, Germany
| | - Twinkle Bhatia
- Department of Biotechnology and Biochemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, Halle, 06120, Saxony-Anhalt, Germany
| | - Bruno Voigt
- Department of Physics, Martin-Luther-University Halle-Wittenberg, Betty-Heimann-Strasse 7, Halle, 06120, Saxony-Anhalt, Germany
| | - Hauke Lilie
- Department of Biotechnology and Biochemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, Halle, 06120, Saxony-Anhalt, Germany
| | - Maria Ott
- Department of Biotechnology and Biochemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, Halle, 06120, Saxony-Anhalt, Germany
| |
Collapse
|
11
|
Kumar R, Arroyo-García LE, Manchanda S, Adam L, Pizzirusso G, Biverstål H, Nilsson P, Fisahn A, Johansson J, Abelein A. Molecular Mechanisms of Amyloid-β Self-Assembly Seeded by In Vivo-Derived Fibrils and Inhibitory Effects of the BRICHOS Chaperone. ACS Chem Neurosci 2023; 14. [PMID: 37023330 PMCID: PMC10119923 DOI: 10.1021/acschemneuro.3c00044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Self-replication of amyloid-β-peptide (Aβ) fibril formation is a hallmark in Alzheimer's disease (AD). Detailed insights have been obtained in Aβ self-assembly in vitro, yet whether similar mechanisms are relevant in vivo has remained elusive. Here, we investigated the ability of in vivo-derived Aβ fibrils from two different amyloid precursor protein knock-in AD mouse models to seed Aβ42 aggregation, where we quantified the microscopic rate constants. We found that the nucleation mechanism of in vivo-derived fibril-seeded Aβ42 aggregation can be described with the same kinetic model as that in vitro. Further, we identified the inhibitory mechanism of the anti-amyloid BRICHOS chaperone on seeded Aβ42 fibrillization, revealing a suppression of secondary nucleation and fibril elongation, which is strikingly similar as observed in vitro. These findings hence provide a molecular understanding of the Aβ42 nucleation process triggered by in vivo-derived Aβ42 propagons, providing a framework for the search for new AD therapeutics.
Collapse
Affiliation(s)
- Rakesh Kumar
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Luis Enrique Arroyo-García
- Division
of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology,
Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Department
of Women’s and Children’s Health, Karolinska Institutet, 171 64 Solna, Sweden
| | - Shaffi Manchanda
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Laurène Adam
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Giusy Pizzirusso
- Division
of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology,
Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
- Department
of Women’s and Children’s Health, Karolinska Institutet, 171 64 Solna, Sweden
| | - Henrik Biverstål
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Per Nilsson
- Division
of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology,
Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - André Fisahn
- Division
of Neurogeriatrics; Center for Alzheimer Research; Department of Neurobiology,
Care Sciences and Society, Karolinska Institutet, 171 64 Solna, Sweden
| | - Jan Johansson
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| | - Axel Abelein
- Department
of Biosciences and Nutrition, Karolinska
Institutet, 141 52 Huddinge, Sweden
| |
Collapse
|
12
|
Sakalauskas A, Ziaunys M, Snieckute R, Janoniene A, Veiveris D, Zvirblis M, Dudutiene V, Smirnovas V. The Major Components of Cerebrospinal Fluid Dictate the Characteristics of Inhibitors against Amyloid-Beta Aggregation. Int J Mol Sci 2023; 24:ijms24065991. [PMID: 36983069 PMCID: PMC10059578 DOI: 10.3390/ijms24065991] [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: 02/27/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
The main pathological hallmark of Alzheimer's disease (AD) is the aggregation of amyloid-β into amyloid fibrils, leading to a neurodegeneration cascade. The current medications are far from sufficient to prevent the onset of the disease, hence requiring more research to find new alternative drugs for curing AD. In vitro inhibition experiments are one of the primary tools in testing whether a molecule may be potent to impede the aggregation of amyloid-beta peptide (Aβ42). However, kinetic experiments in vitro do not match the mechanism found when aggregating Aβ42 in cerebrospinal fluid. The different aggregation mechanisms and the composition of the reaction mixtures may also impact the characteristics of the inhibitor molecules. For this reason, altering the reaction mixture to resemble components found in cerebrospinal fluid (CSF) is critical to partially compensate for the mismatch between the inhibition experiments in vivo and in vitro. In this study, we used an artificial cerebrospinal fluid that contained the major components found in CSF and performed Aβ42 aggregation inhibition studies using oxidized epigallocatechin-3-gallate (EGCG) and fluorinated benzenesulfonamide VR16-09. This led to a discovery of a complete turnaround of their inhibitory characteristics, rendering EGCG ineffective while significantly improving the efficacy of VR16-09. HSA was the main contributor in the mixture that significantly increased the anti-amyloid characteristics of VR16-09.
Collapse
Affiliation(s)
- Andrius Sakalauskas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Mantas Ziaunys
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Ruta Snieckute
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Agne Janoniene
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Dominykas Veiveris
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Mantas Zvirblis
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Virginija Dudutiene
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania
| |
Collapse
|
13
|
Ghosh S, Ali R, Verma S. Aβ-oligomers: A potential therapeutic target for Alzheimer's disease. Int J Biol Macromol 2023; 239:124231. [PMID: 36996958 DOI: 10.1016/j.ijbiomac.2023.124231] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023]
Abstract
The cascade of amyloid formation relates to multiple complex events at the molecular level. Previous research has established amyloid plaque deposition as the leading cause of Alzheimer's disease (AD) pathogenesis, detected mainly in aged population. The primary components of the plaques are two alloforms of amyloid-beta (Aβ), Aβ1-42 and Aβ1-40 peptides. Recent studies have provided considerable evidence contrary to the previous claim indicating that amyloid-beta oligomers (AβOs) as the main culprit responsible for AD-associated neurotoxicity and pathogenesis. In this review, we have discussed the primary features of AβOs, such as assembly formation, the kinetics of oligomer formation, interactions with various membranes/membrane receptors, the origin of toxicity, and oligomer-specific detection methods. Recently, the discovery of rationally designed antibodies has opened a gateway for using synthesized peptides as a grafting component in the complementarity determining region (CDR) of antibodies. Thus, the Aβ sequence motif or the complementary peptide sequence in the opposite strand of the β-sheet (extracted from the Protein Data Bank: PDB) helps design oligomer-specific inhibitors. The microscopic event responsible for oligomer formation can be targeted, and thus prevention of the overall macroscopic behaviour of the aggregation or the associated toxicity can be achieved. We have carefully reviewed the oligomer formation kinetics and associated parameters. Besides, we have depicted a thorough understanding of how the synthesized peptide inhibitors can impede the early aggregates (oligomers), mature fibrils, monomers, or a mixture of the species. The oligomer-specific inhibitors (peptides or peptide fragments) lack in-depth chemical kinetics and optimization control-based screening. In the present review, we have proposed a hypothesis for effectively screening oligomer-specific inhibitors using the chemical kinetics (determining the kinetic parameters) and optimization control strategy (cost-dependent analysis). Further, it may be possible to implement the structure-kinetic-activity-relationship (SKAR) strategy instead of structure-activity-relationship (SAR) to improve the inhibitor's activity. The controlled optimization of the kinetic parameters and dose usage will be beneficial for narrowing the search window for the inhibitors.
Collapse
|
14
|
Nguyen PH, Sterpone F, Derreumaux P. Self-Assembly of Amyloid-Beta (Aβ) Peptides from Solution to Near In Vivo Conditions. J Phys Chem B 2022; 126:10317-10326. [PMID: 36469912 DOI: 10.1021/acs.jpcb.2c06375] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding the atomistic resolution changes during the self-assembly of amyloid peptides or proteins is important to develop compounds or conditions to alter the aggregation pathways and suppress the toxicity and potentially aid in the development of drugs. However, the complexity of protein aggregation and the transient order/disorder of oligomers along the pathways to fibril are very challenging. In this Perspective, we discuss computational studies of amyloid polypeptides carried out under various conditions, including conditions closely mimicking in vivo and point out the challenges in obtaining physiologically relevant results, focusing mainly on the amyloid-beta Aβ peptides.
Collapse
Affiliation(s)
- Phuong H Nguyen
- CNRS, Université Paris Cité, UPR 9080, Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, 13 rue Pierre et Marie Curie, 75005 Paris, France
| | - Fabio Sterpone
- CNRS, Université Paris Cité, UPR 9080, Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, 13 rue Pierre et Marie Curie, 75005 Paris, France
| | - Philippe Derreumaux
- CNRS, Université Paris Cité, UPR 9080, Laboratoire de Biochimie Théorique, Institut de Biologie Physico-Chimique, Fondation Edmond de Rothschild, 13 rue Pierre et Marie Curie, 75005 Paris, France.,Institut Universitaire de France (IUF), 75005, Paris, France
| |
Collapse
|
15
|
Taylor AP, Davis PJ, Aubrey LD, White JBR, Parton ZN, Staniforth RA. Simple, Reliable Protocol for High-Yield Solubilization of Seedless Amyloid-β Monomer. ACS Chem Neurosci 2022; 14:53-71. [PMID: 36512740 PMCID: PMC9817077 DOI: 10.1021/acschemneuro.2c00411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Self-assembly of the amyloid-β (Aβ) peptide to form toxic oligomers and fibrils is a key causal event in the onset of Alzheimer's disease, and Aβ is the focus of intense research in neuroscience, biophysics, and structural biology aimed at therapeutic development. Due to its rapid self-assembly and extreme sensitivity to aggregation conditions, preparation of seedless, reproducible Aβ solutions is highly challenging, and there are serious ongoing issues with consistency in the literature. In this paper, we use a liquid-phase separation technique, asymmetric flow field-flow fractionation with multiangle light scattering (AF4-MALS), to develop and validate a simple, effective, economical method for re-solubilization and quality control of purified, lyophilized Aβ samples. Our findings were obtained with recombinant peptide but are physicochemical in nature and thus highly relevant to synthetic peptide. We show that much of the variability in the literature stems from the inability of overly mild solvent treatments to produce consistently monomeric preparations and is rectified by a protocol involving high-pH (>12) dissolution, sonication, and rapid freezing to prevent modification. Aβ treated in this manner is chemically stable, can be stored over long timescales at -80 °C, and exhibits remarkably consistent self-assembly behavior when returned to near-neutral pH. These preparations are highly monomeric, seedless, and do not require additional rounds of size exclusion, eliminating the need for this costly procedure and increasing the flexibility of use. We propose that our improved protocol is the simplest, fastest, and most effective way to solubilize Aβ from diverse sources for sensitive self-assembly and toxicity assays.
Collapse
|
16
|
Thacker D, Willas A, Dear AJ, Linse S. Role of Hydrophobicity at the N-Terminal Region of Aβ42 in Secondary Nucleation. ACS Chem Neurosci 2022; 13:3477-3487. [PMID: 36411082 PMCID: PMC9732875 DOI: 10.1021/acschemneuro.2c00504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/19/2022] [Indexed: 11/23/2022] Open
Abstract
The self-assembly of the amyloid β 42 (Aβ42) peptide is linked to Alzheimer's disease, and oligomeric intermediates are linked to neuronal cell death during the pathology of the disease. These oligomers are produced prolifically during secondary nucleation, by which the aggregation of monomers is catalyzed on fibril surfaces. Significant progress has been made in understanding the aggregation mechanism of Aβ42; still, a detailed molecular-level understanding of secondary nucleation is lacking. Here, we explore the role of four hydrophobic residues on the unstructured N-terminal region of Aβ42 in secondary nucleation. We create eight mutants with single substitutions at one of the four positions─Ala2, Phe4, Tyr10, and Val12─to decrease the hydrophobicity at respective positions (A2T, A2S, F4A, F4S, Y10A, Y10S, V12A, and V12S) and one mutant (Y10F) to remove the polar nature of Tyr10. Kinetic analyses of aggregation data reveal that the hydrophobicity at the N-terminal region of Aβ42, especially at positions 10 and 12, affects the rate of fibril mass generated via secondary nucleation. Cryo-electron micrographs reveal that most of the mutants with lower hydrophobicity form fibrils that are markedly longer than WT Aβ42, in line with the reduced secondary nucleation rates for these peptides. The dominance of secondary nucleation, however, is still retained in the aggregation mechanism of these mutants because the rate of primary nucleation is even more reduced. This highlights that secondary nucleation is a general phenomenon that is not dependent on any one particular feature of the peptide and is rather robust to sequence perturbations.
Collapse
Affiliation(s)
- Dev Thacker
- Department
of Biochemistry and Structural Biology, Lund University, Lund22362, Sweden
| | - Amanda Willas
- Department
of Biochemistry and Structural Biology, Lund University, Lund22362, Sweden
| | - Alexander J. Dear
- Department
of Biochemistry and Structural Biology, Lund University, Lund22362, Sweden
- Centre
for Misfolding Diseases, Department of Chemistry, University of Cambridge, CambridgeCB2 1EW, U.K.
| | - Sara Linse
- Department
of Biochemistry and Structural Biology, Lund University, Lund22362, Sweden
| |
Collapse
|
17
|
McMackin P, Adam J, Griffin S, Hirsa A. Amyloidogenesis via interfacial shear in a containerless biochemical reactor aboard the International Space Station. NPJ Microgravity 2022; 8:41. [PMID: 36127358 PMCID: PMC9489778 DOI: 10.1038/s41526-022-00227-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022] Open
Abstract
Fluid interfaces significantly influence the dynamics of protein solutions, effects that can be isolated by performing experiments in microgravity, greatly reducing the amount of solid boundaries present, allowing air-liquid interfaces to become dominant. This investigation examined the effects of protein concentration on interfacial shear-induced fibrillization of insulin in microgravity within a containerless biochemical reactor, the ring-sheared drop (RSD), aboard the international space station (ISS). Human insulin was used as a model amyloidogenic protein for studying protein kinetics with applications to in situ pharmaceutical production, tissue engineering, and diseases such as Alzheimer’s, Parkinson’s, infectious prions, and type 2 diabetes. Experiments investigated three main stages of amyloidogenesis: nucleation studied by seeding native solutions with fibril aggregates, fibrillization quantified using intrinsic fibrillization rate after fitting measured solution intensity to a sigmoidal function, and gelation observed by detection of solidification fronts. Results demonstrated that in surface-dominated amyloidogenic protein solutions: seeding with fibrils induces fibrillization of native protein, intrinsic fibrillization rate is independent of concentration, and that there is a minimum fibril concentration for gelation with gelation rate and rapidity of onset increasing monotonically with increasing protein concentration. These findings matched well with results of previous studies within ground-based analogs.
Collapse
Affiliation(s)
- Patrick McMackin
- Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA
| | - Joe Adam
- Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA.,Department of Biological Sciences, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA.,Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA
| | - Shannon Griffin
- Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA.,Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA
| | - Amir Hirsa
- Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA. .,Chemical and Biological Engineering, Rensselaer Polytechnic Institute, 110 8th St, Troy, 12180, NY, USA.
| |
Collapse
|
18
|
Weiffert T, Meisl G, Curk S, Cukalevski R, Šarić A, Knowles TPJ, Linse S. Influence of denaturants on amyloid β42 aggregation kinetics. Front Neurosci 2022; 16:943355. [PMID: 36203800 PMCID: PMC9531139 DOI: 10.3389/fnins.2022.943355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
Amyloid formation is linked to devastating neurodegenerative diseases, motivating detailed studies of the mechanisms of amyloid formation. For Aβ, the peptide associated with Alzheimer's disease, the mechanism and rate of aggregation have been established for a range of variants and conditions in vitro and in bodily fluids. A key outstanding question is how the relative stabilities of monomers, fibrils and intermediates affect each step in the fibril formation process. By monitoring the kinetics of aggregation of Aβ42, in the presence of urea or guanidinium hydrochloride (GuHCl), we here determine the rates of the underlying microscopic steps and establish the importance of changes in relative stability induced by the presence of denaturant for each individual step. Denaturants shift the equilibrium towards the unfolded state of each species. We find that a non-ionic denaturant, urea, reduces the overall aggregation rate, and that the effect on nucleation is stronger than the effect on elongation. Urea reduces the rate of secondary nucleation by decreasing the coverage of fibril surfaces and the rate of nucleus formation. It also reduces the rate of primary nucleation, increasing its reaction order. The ionic denaturant, GuHCl, accelerates the aggregation at low denaturant concentrations and decelerates the aggregation at high denaturant concentrations. Below approximately 0.25 M GuHCl, the screening of repulsive electrostatic interactions between peptides by the charged denaturant dominates, leading to an increased aggregation rate. At higher GuHCl concentrations, the electrostatic repulsion is completely screened, and the denaturing effect dominates. The results illustrate how the differential effects of denaturants on stability of monomer, oligomer and fibril translate to differential effects on microscopic steps, with the rate of nucleation being most strongly reduced.
Collapse
Affiliation(s)
- Tanja Weiffert
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Georg Meisl
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Samo Curk
- Department of Physics and Astronomy, University College London, London, United Kingdom
| | - Risto Cukalevski
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| | - Anđela Šarić
- Department of Physics and Astronomy, University College London, London, United Kingdom
| | - Tuomas P. J. Knowles
- Yusuf Hamied Department of Chemistry, Centre for Misfolding Diseases, University of Cambridge, Cambridge, United Kingdom
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - Sara Linse
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
| |
Collapse
|
19
|
Meisl G, Xu CK, Taylor JD, Michaels TCT, Levin A, Otzen D, Klenerman D, Matthews S, Linse S, Andreasen M, Knowles TPJ. Uncovering the universality of self-replication in protein aggregation and its link to disease. SCIENCE ADVANCES 2022; 8:eabn6831. [PMID: 35960802 PMCID: PMC9374340 DOI: 10.1126/sciadv.abn6831] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Fibrillar protein aggregates are a hallmark of a range of human disorders, from prion diseases to dementias, but are also encountered in several functional contexts. Yet, the fundamental links between protein assembly mechanisms and their functional or pathological roles have remained elusive. Here, we analyze the aggregation kinetics of a large set of proteins that self-assemble by a nucleated-growth mechanism, from those associated with disease, over those whose aggregates fulfill functional roles in biology, to those that aggregate only under artificial conditions. We find that, essentially, all such systems, regardless of their biological role, are capable of self-replication. However, for aggregates that have evolved to fulfill a structural role, the rate of self-replication is too low to be significant on the biologically relevant time scale. By contrast, all disease-related proteins are able to self-replicate quickly compared to the time scale of the associated disease. Our findings establish the ubiquity of self-replication and point to its potential importance across aggregation-related disorders.
Collapse
Affiliation(s)
- Georg Meisl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Catherine K. Xu
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Jonathan D. Taylor
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Thomas C. T. Michaels
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Aviad Levin
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - Daniel Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, Aarhus DK-8000, Denmark
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- U.K. Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK
| | - Steve Matthews
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Sara Linse
- Department of Biochemistry and Structural Biology, Lund University, Lund, Sweden
- Corresponding author. (S.L.); (M.A.); (T.P.J.K.)
| | - Maria Andreasen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- Department of Biomedicine, Aarhus University, Wilhelm Meyers Allé 3, Aarhus DK-8000, Denmark
- Corresponding author. (S.L.); (M.A.); (T.P.J.K.)
| | - Tuomas P. J. Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- Cavendish Laboratory, University of Cambridge, 19 JJ Thomson Avenue, Cambridge CB3 0HE, UK
- Corresponding author. (S.L.); (M.A.); (T.P.J.K.)
| |
Collapse
|
20
|
Meisl G, Knowles TPJ, Klenerman D. Mechanistic Models of Protein Aggregation Across Length-Scales and Time-Scales: From the Test Tube to Neurodegenerative Disease. Front Neurosci 2022; 16:909861. [PMID: 35844223 PMCID: PMC9281552 DOI: 10.3389/fnins.2022.909861] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/31/2022] [Indexed: 11/29/2022] Open
Abstract
Through advances in the past decades, the central role of aberrant protein aggregation has been established in many neurodegenerative diseases. Crucially, however, the molecular mechanisms that underlie aggregate proliferation in the brains of affected individuals are still only poorly understood. Under controlled in vitro conditions, significant progress has been made in elucidating the molecular mechanisms that take place during the assembly of purified protein molecules, through advances in both experimental methods and the theories used to analyse the resulting data. The determination of the aggregation mechanism for a variety of proteins revealed the importance of intermediate oligomeric species and of the interactions with promotors and inhibitors. Such mechanistic insights, if they can be achieved in a disease-relevant system, provide invaluable information to guide the design of potential cures to these devastating disorders. However, as experimental systems approach the situation present in real disease, their complexity increases substantially. Timescales increase from hours an aggregation reaction takes in vitro, to decades over which the process takes place in disease, and length-scales increase to the dimension of a human brain. Thus, molecular level mechanistic studies, like those that successfully determined mechanisms in vitro, have only been applied in a handful of living systems to date. If their application can be extended to further systems, including patient data, they promise powerful new insights. Here we present a review of the existing strategies to gain mechanistic insights into the molecular steps driving protein aggregation and discuss the obstacles and potential paths to achieving their application in disease. First, we review the experimental approaches and analysis techniques that are used to establish the aggregation mechanisms in vitro and the insights that have been gained from them. We then discuss how these approaches must be modified and adapted to be applicable in vivo and review the existing works that have successfully applied mechanistic analysis of protein aggregation in living systems. Finally, we present a broad mechanistic classification of in vivo systems and discuss what will be required to further our understanding of aggregate formation in living systems.
Collapse
Affiliation(s)
- Georg Meisl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Tuomas P. J. Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
- UK Dementia Research Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
21
|
Taylor AIP, Staniforth RA. General Principles Underpinning Amyloid Structure. Front Neurosci 2022; 16:878869. [PMID: 35720732 PMCID: PMC9201691 DOI: 10.3389/fnins.2022.878869] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/11/2022] [Indexed: 12/14/2022] Open
Abstract
Amyloid fibrils are a pathologically and functionally relevant state of protein folding, which is generally accessible to polypeptide chains and differs fundamentally from the globular state in terms of molecular symmetry, long-range conformational order, and supramolecular scale. Although amyloid structures are challenging to study, recent developments in techniques such as cryo-EM, solid-state NMR, and AFM have led to an explosion of information about the molecular and supramolecular organization of these assemblies. With these rapid advances, it is now possible to assess the prevalence and significance of proposed general structural features in the context of a diverse body of high-resolution models, and develop a unified view of the principles that control amyloid formation and give rise to their unique properties. Here, we show that, despite system-specific differences, there is a remarkable degree of commonality in both the structural motifs that amyloids adopt and the underlying principles responsible for them. We argue that the inherent geometric differences between amyloids and globular proteins shift the balance of stabilizing forces, predisposing amyloids to distinct molecular interaction motifs with a particular tendency for massive, lattice-like networks of mutually supporting interactions. This general property unites previously characterized structural features such as steric and polar zippers, and contributes to the long-range molecular order that gives amyloids many of their unique properties. The shared features of amyloid structures support the existence of shared structure-activity principles that explain their self-assembly, function, and pathogenesis, and instill hope in efforts to develop broad-spectrum modifiers of amyloid function and pathology.
Collapse
|
22
|
Chatterjee D, Jacob RS, Ray S, Navalkar A, Singh N, Sengupta S, Gadhe L, Kadu P, Datta D, Paul A, Arunima S, Mehra S, Pindi C, Kumar S, Singru P, Senapati S, Maji SK. Co-aggregation and secondary nucleation in the life cycle of human prolactin/galanin functional amyloids. eLife 2022; 11:73835. [PMID: 35257659 PMCID: PMC8993219 DOI: 10.7554/elife.73835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 03/03/2022] [Indexed: 11/22/2022] Open
Abstract
Synergistic-aggregation and cross-seeding by two different proteins/peptides in the amyloid aggregation are well evident in various neurological disorders including Alzheimer’s disease. Here, we show co-storage of human Prolactin (PRL), which is associated with lactation in mammals, and neuropeptide galanin (GAL) as functional amyloids in secretory granules (SGs) of the female rat. Using a wide variety of biophysical studies, we show that irrespective of the difference in sequence and structure, both hormones facilitate their synergic aggregation to amyloid fibrils. Although each hormone possesses homotypic seeding ability, a unidirectional cross-seeding of GAL aggregation by PRL seeds and the inability of cross seeding by mixed fibrils suggest tight regulation of functional amyloid formation by these hormones for their efficient storage in SGs. Further, the faster release of functional hormones from mixed fibrils compared to the corresponding individual amyloid, suggests a novel mechanism of heterologous amyloid formation in functional amyloids of SGs in the pituitary. The formation of plaques of proteins called ‘amyloids’ in the brain is one of the hallmark characteristics of both Alzheimer’s and Parkinson’s disease, but amyloids can form in many tissues and organs, often disrupting normal activity. A lot of the research into amyloids has focused on their role in disease, but it turns out that amyloids can also appear in healthy tissues. For example, some protein hormones form amyloids that act as storage depots, helping cells to release the hormone when it is needed. Normally, amyloids are made mostly of a single type of protein or protein fragment associated with a particular disease like Alzheimer's. Often, this type of amyloid promotes plaque formation in other proteins, which aggravates other diseases (for example, the amyloids that form in Alzheimer’s can lead to Parkinson’s disease or type II diabetes getting worse).The plaques start growing from small amyloid fragments called seeds. In mixed amyloids – amyloids made of two types of proteins – seeds made of one protein can trigger the formation of amyloids of the other protein. This raises the question, is this true for hormones? The body often releases more than one hormone at a time from the same tissue; for example, the pituitary gland releases prolactin and galanin simultaneously. However, these hormones have completely different structures, so whether they can form a mixed amyloid is unclear. To answer this question, Chatterjee et al. first determined that, within the pituitary gland of female rats, prolactin and galanin could be found together in the same cells, forming mixed amyloids. To understand out how this happens, Chatterjee et al. tried seeding new amyloids using either prolactin or galanin. This revealed that only prolactin seeds were able to trigger the formation of galanin amyloids. Chatterjee et al. also found that the mixed amyloids could release the hormones faster than amyloids made from either protein alone. Together, these results suggest that the collaboration between these two proteins may help maintain hormone balance in the body. Problems with hormone storage and release lead to various human diseases, including prolactinoma. Understanding amyloid storage depots could reveal new ways to control hormone levels. Further research could also help to explain more about well-studied diseases linked to amyloids, like Alzheimer's.
Collapse
Affiliation(s)
- Debdeep Chatterjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Reeba S Jacob
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Soumik Ray
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Namrata Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Shinjinee Sengupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Laxmikant Gadhe
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Debalina Datta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ajoy Paul
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Sakunthala Arunima
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Surabhi Mehra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Chinmai Pindi
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Santosh Kumar
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
| | - Praful Singru
- School of Biological Sciences, National Institute of Science Education and Research, Bhubaneswar, India
| | - Sanjib Senapati
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Samir K Maji
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
23
|
Braun GA, Dear AJ, Sanagavarapu K, Zetterberg H, Linse S. Amyloid-β peptide 37, 38 and 40 individually and cooperatively inhibit amyloid-β 42 aggregation. Chem Sci 2022; 13:2423-2439. [PMID: 35310497 PMCID: PMC8864715 DOI: 10.1039/d1sc02990h] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 01/22/2022] [Indexed: 12/22/2022] Open
Abstract
The pathology of Alzheimer's disease is connected to the aggregation of β-amyloid (Aβ) peptide, which in vivo exists as a number of length-variants. Truncations and extensions are found at both the N- and C-termini, relative to the most commonly studied 40- and 42-residue alloforms. Here, we investigate the aggregation of two physiologically abundant alloforms, Aβ37 and Aβ38, as pure peptides and in mixtures with Aβ40 and Aβ42. A variety of molar ratios were applied in quaternary mixtures to investigate whether a certain ratio is maximally inhibiting of the more toxic alloform Aβ42. Through kinetic analysis, we show that both Aβ37 and Aβ38 self-assemble through an autocatalytic secondary nucleation reaction to form fibrillar β-sheet-rich aggregates, albeit on a longer timescale than Aβ40 or Aβ42. Additionally, we show that the shorter alloforms co-aggregate with Aβ40, affecting both the kinetics of aggregation and the resulting fibrillar ultrastructure. In contrast, neither Aβ37 nor Aβ38 forms co-aggregates with Aβ42; however, both short alloforms reduce the rate of Aβ42 aggregation in a concentration-dependent manner. Finally, we show that the aggregation of Aβ42 is more significantly impeded by a combination of Aβ37, Aβ38, and Aβ40 than by any of these alloforms independently. These results demonstrate that the aggregation of any given Aβ alloform is significantly perturbed by the presence of other alloforms, particularly in heterogeneous mixtures, such as is found in the extracellular fluid of the brain.
Collapse
Affiliation(s)
- Gabriel A Braun
- Biochemistry and Structural Biology, Lund University Lund Sweden
| | - Alexander J Dear
- Biochemistry and Structural Biology, Lund University Lund Sweden
- Department of Cell Biology, Harvard Medical School Boston MA USA
- Paulson School of Engineering and Applied Science, Harvard University Cambridge MA USA
- Department of Chemistry, University of Cambridge Cambridge UK
| | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg Mölndal Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital Mölndal Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology Queen Square London UK
- UK Dementia Research Institute at UCL London UK
| | - Sara Linse
- Biochemistry and Structural Biology, Lund University Lund Sweden
| |
Collapse
|
24
|
Zimmermann MR, Bera SC, Meisl G, Dasadhikari S, Ghosh S, Linse S, Garai K, Knowles TPJ. Mechanism of Secondary Nucleation at the Single Fibril Level from Direct Observations of Aβ42 Aggregation. J Am Chem Soc 2021; 143:16621-16629. [PMID: 34582216 DOI: 10.1021/jacs.1c07228] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The formation of amyloid fibrils and oligomers is a hallmark of several neurodegenerative disorders, including Alzheimer's disease (AD), and contributes to the disease pathway. To progress our understanding of these diseases at a molecular level, it is crucial to determine the mechanisms and rates of amyloid formation and replication. In the context of AD, the self-replication of aggregates of the Aβ42 peptide by secondary nucleation, leading to the formation of new aggregates on the surfaces of existing ones, is a major source of both new fibrils and smaller toxic oligomeric species. However, the core mechanistic determinants, including the presence of intermediates, as well as the role of heterogeneities in the fibril population, are challenging to determine from bulk aggregation measurements. Here, we obtain such information by monitoring directly the time evolution of individual fibrils by TIRF microscopy. Crucially, essentially all aggregates have the ability to self-replicate via secondary nucleation, and the amplification of the aggregate concentration cannot be explained by a small fraction of "superspreader" fibrils. We observe that secondary nucleation is a catalytic multistep process involving the attachment of soluble species to the fibril surface, followed by conversion/detachment to yield a new fibril in solution. Furthermore, we find that fibrils formed by secondary nucleation resemble the parent fibril population. This detailed level of mechanistic insights into aggregate self-replication is key in the rational design of potential inhibitors of this process.
Collapse
Affiliation(s)
- Manuela R Zimmermann
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | - Subhas C Bera
- TIFR Centre for Interdisciplinary Sciences, 500046 Hyderabad, India
- Interdisciplinary Center for Clinical Research, Friedrich-Alexander-University, Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Georg Meisl
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
| | | | - Shamasree Ghosh
- TIFR Centre for Interdisciplinary Sciences, 500046 Hyderabad, India
| | - Sara Linse
- Department of Chemistry, Division for Biochemistry and Structural Biology, Lund University, 221 00 Lund, Sweden
| | - Kanchan Garai
- TIFR Centre for Interdisciplinary Sciences, 500046 Hyderabad, India
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, United Kingdom
- Cavendish Laboratory, University of Cambridge, CB3 0HE Cambridge, United Kingdom
| |
Collapse
|
25
|
Adam JA, Middlestead HR, Debono NE, Hirsa AH. Effects of Shear Rate and Protein Concentration on Amyloidogenesis via Interfacial Shear. J Phys Chem B 2021; 125:10355-10363. [PMID: 34478304 DOI: 10.1021/acs.jpcb.1c05171] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The influence of hydrodynamics on protein fibrillization kinetics is relevant to biophysics, biochemical reactors, medicine, and disease. This investigation focused on the effects of interfacial shear on the fibrillization kinetics of insulin. Human insulin served as a model protein for studying shear-induced fibrillization with relevance to amyloid diseases such as Alzheimer's, Parkinson's, prions, and type 2 diabetes. Insulin solutions at different protein concentrations were subjected to shear flows with prescribed interfacial angular velocities using a knife-edge (surface) viscometer (KEV) operating in a laminar axisymmetric flow regime where inertia is significant. Fibrillization kinetics were quantified using intrinsic fibrillization rate and times (onset, half, and end) determined through spectroscopic measurement of monomer extinction curves and fitting to a sigmoidal function. Additionally, the occurrence of gelation was determined through macroscopic imaging and transient fibril microstructure was captured using fluorescence microscopy. The results showed that increasing interfacial shear rate produced a monotonic increase in intrinsic fibrillization rate and a monotonic decrease in fibrillization time. Protein concentration did not significantly impact the intrinsic fibrillization rate or times; however, a minimum fibril concentration for gelation was found. Protein microstructure showed increasing aggregation and plaque/cluster formation with time.
Collapse
Affiliation(s)
| | - Hannah R Middlestead
- Chemical Engineering, University of Colorado Boulder, Boulder, Colorado 80309-0584, United States
| | | | | |
Collapse
|
26
|
Sakalauskas A, Ziaunys M, Snieckute R, Smirnovas V. Autoxidation Enhances Anti-Amyloid Potential of Flavone Derivatives. Antioxidants (Basel) 2021; 10:antiox10091428. [PMID: 34573060 PMCID: PMC8465893 DOI: 10.3390/antiox10091428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
The increasing prevalence of amyloid-related disorders, such as Alzheimer's or Parkinson's disease, raises the need for effective anti-amyloid drugs. It has been shown on numerous occasions that flavones, a group of naturally occurring anti-oxidants, can impact the aggregation process of several amyloidogenic proteins and peptides, including amyloid-beta. Due to flavone autoxidation at neutral pH, it is uncertain if the effective inhibitor is the initial molecule or a product of this reaction, as many anti-amyloid assays attempt to mimic physiological conditions. In this work, we examine the aggregation-inhibiting properties of flavones before and after they are oxidized. The oxidation of flavones was monitored by measuring the UV-vis absorbance spectrum change over time. The protein aggregation kinetics were followed by measuring the amyloidophilic dye thioflavin-T (ThT) fluorescence intensity change. Atomic force microscopy was employed to image the aggregates formed with the most prominent inhibitors. We demonstrate that flavones, which undergo autoxidation, have a far greater potency at inhibiting the aggregation of both the disease-related amyloid-beta, as well as a model amyloidogenic protein-insulin. Oxidized 6,2',3'-trihydroxyflavone was the most potent inhibitor affecting both insulin (7-fold inhibition) and amyloid-beta (2-fold inhibition). We also show that this tendency to autoxidize is related to the positions of the flavone hydroxyl groups.
Collapse
|
27
|
Rahman MM, Lendel C. Extracellular protein components of amyloid plaques and their roles in Alzheimer's disease pathology. Mol Neurodegener 2021; 16:59. [PMID: 34454574 PMCID: PMC8400902 DOI: 10.1186/s13024-021-00465-0] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is pathologically defined by the presence of fibrillar amyloid β (Aβ) peptide in extracellular senile plaques and tau filaments in intracellular neurofibrillary tangles. Extensive research has focused on understanding the assembly mechanisms and neurotoxic effects of Aβ during the last decades but still we only have a brief understanding of the disease associated biological processes. This review highlights the many other constituents that, beside Aβ, are accumulated in the plaques, with the focus on extracellular proteins. All living organisms rely on a delicate network of protein functionality. Deposition of significant amounts of certain proteins in insoluble inclusions will unquestionably lead to disturbances in the network, which may contribute to AD and copathology. This paper provide a comprehensive overview of extracellular proteins that have been shown to interact with Aβ and a discussion of their potential roles in AD pathology. Methods that can expand the knowledge about how the proteins are incorporated in plaques are described. Top-down methods to analyze post-mortem tissue and bottom-up approaches with the potential to provide molecular insights on the organization of plaque-like particles are compared. Finally, a network analysis of Aβ-interacting partners with enriched functional and structural key words is presented.
Collapse
Affiliation(s)
- M Mahafuzur Rahman
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden.
| | - Christofer Lendel
- Department of Chemistry, KTH Royal Institute of Technology, SE-100 44, Stockholm, Sweden.
| |
Collapse
|
28
|
Thompson TB, Meisl G, Knowles TPJ, Goriely A. The role of clearance mechanisms in the kinetics of pathological protein aggregation involved in neurodegenerative diseases. J Chem Phys 2021; 154:125101. [PMID: 33810689 DOI: 10.1063/5.0031650] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The deposition of pathological protein aggregates in the brain plays a central role in cognitive decline and structural damage associated with neurodegenerative diseases. In Alzheimer's disease, the formation of amyloid-β plaques and neurofibrillary tangles of the tau protein is associated with the appearance of symptoms and pathology. Detailed models for the specific mechanisms of aggregate formation, such as nucleation and elongation, exist for aggregation in vitro where the total protein mass is conserved. However, in vivo, an additional class of mechanisms that clear pathological species is present and is believed to play an essential role in limiting the formation of aggregates and preventing or delaying the emergence of disease. A key unanswered question in the field of neuro-degeneration is how these clearance mechanisms can be modeled and how alterations in the processes of clearance or aggregation affect the stability of the system toward aggregation. Here, we generalize classical models of protein aggregation to take into account both production of monomers and the clearance of protein aggregates. We show that, depending on the specifics of the clearance process, a critical clearance value emerges above which accumulation of aggregates does not take place. Our results show that a sudden switch from a healthy to a disease state can be caused by small variations in the efficiency of the clearance process and provide a mathematical framework to explore the detailed effects of different mechanisms of clearance on the accumulation of aggregates.
Collapse
Affiliation(s)
- T B Thompson
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - G Meisl
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - T P J Knowles
- Department of Chemistry and Department of Physics, University of Cambridge, Cambridge, United Kingdom
| | - A Goriely
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
29
|
Willbold D, Strodel B, Schröder GF, Hoyer W, Heise H. Amyloid-type Protein Aggregation and Prion-like Properties of Amyloids. Chem Rev 2021; 121:8285-8307. [PMID: 34137605 DOI: 10.1021/acs.chemrev.1c00196] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review will focus on the process of amyloid-type protein aggregation. Amyloid fibrils are an important hallmark of protein misfolding diseases and therefore have been investigated for decades. Only recently, however, atomic or near-atomic resolution structures have been elucidated from various in vitro and ex vivo obtained fibrils. In parallel, the process of fibril formation has been studied in vitro under highly artificial but comparatively reproducible conditions. The review starts with a summary of what is known and speculated from artificial in vitro amyloid-type protein aggregation experiments. A partially hypothetic fibril selection model will be described that may be suitable to explain why amyloid fibrils look the way they do, in particular, why at least all so far reported high resolution cryo-electron microscopy obtained fibril structures are in register, parallel, cross-β-sheet fibrils that mostly consist of two protofilaments twisted around each other. An intrinsic feature of the model is the prion-like nature of all amyloid assemblies. Transferring the model from the in vitro point of view to the in vivo situation is not straightforward, highly hypothetic, and leaves many open questions that need to be addressed in the future.
Collapse
Affiliation(s)
- Dieter Willbold
- Institute of Biological Information Processing, Structural Biochemistry, IBI-7, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany.,Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology (State University), 141700 Dolgoprudny, Russia
| | - Birgit Strodel
- Institute of Biological Information Processing, Structural Biochemistry, IBI-7, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Institute of Theoretical and Computational Chemistry, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Gunnar F Schröder
- Institute of Biological Information Processing, Structural Biochemistry, IBI-7, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Physics Department, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Wolfgang Hoyer
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Henrike Heise
- Institute of Biological Information Processing, Structural Biochemistry, IBI-7, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany.,Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
30
|
Toward the equilibrium and kinetics of amyloid peptide self-assembly. Curr Opin Struct Biol 2021; 70:87-98. [PMID: 34153659 DOI: 10.1016/j.sbi.2021.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/09/2021] [Accepted: 05/09/2021] [Indexed: 01/28/2023]
Abstract
Several devastating human diseases are linked to peptide self-assembly, but our understanding their onset and progression is not settled. This is a sign of the complexity of the aggregation process, which is prevented, catalyzed, or retarded by numerous factors in body fluids and cells, varying in time and space. Biophysical studies of pure peptide solutions contribute insights into the underlying steps in the process and quantitative parameters relating to rate constants (energy barriers) and equilibrium constants (population distributions). This requires methods to quantify the concentration of at least one species in the process. Translation to an in vivo situation poses an enormous challenge, and the effects of selected components (bottom up) or entire body fluids (top down) need to be quantified.
Collapse
|
31
|
Pappas CG, Liu B, Marić I, Ottelé J, Kiani A, van der Klok ML, Onck PR, Otto S. Two Sides of the Same Coin: Emergence of Foldamers and Self-Replicators from Dynamic Combinatorial Libraries. J Am Chem Soc 2021; 143:7388-7393. [PMID: 33955219 PMCID: PMC8154527 DOI: 10.1021/jacs.1c00788] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
The ability of molecules
and systems to make copies of themselves
and the ability of molecules to fold into stable, well-defined three-dimensional
conformations are of considerable importance in the formation and
persistence of life. The question of how, during the emergence of
life, oligomerization reactions become selective and channel these
reactions toward a small number of specific products remains largely
unanswered. Herein, we demonstrate a fully synthetic chemical system
where structurally complex foldamers and self-replicating assemblies
emerge spontaneously and with high selectivity from pools of oligomers
as a result of forming noncovalent interactions. Whether foldamers
or replicators form depends on remarkably small differences in building
block structures and composition and experimental conditions. We also
observed the dynamic transformation of a foldamer into a replicator.
These results show that the structural requirements/design criteria
for building blocks that lead to foldamers are similar to those that
lead to replicators. What determines whether folding or replication
takes place is not necessarily the type of noncovalent interaction,
but only whether they occur intra- or intermolecularly. This work
brings together, for the first time, the fields of replicator and
foldamer chemistry.
Collapse
Affiliation(s)
- Charalampos G Pappas
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Bin Liu
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Ivana Marić
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Jim Ottelé
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Armin Kiani
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Marcus L van der Klok
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Patrick R Onck
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Sijbren Otto
- Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| |
Collapse
|
32
|
Sanfilippo C, Musumeci G, Kazakova M, Mazzone V, Castrogiovanni P, Imbesi R, Di Rosa M. GNG13 Is a Potential Marker of the State of Health of Alzheimer's Disease Patients' Cerebellum. J Mol Neurosci 2021; 71:1046-1060. [PMID: 33057964 DOI: 10.1007/s12031-020-01726-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Brain regions such as the cerebellum (CB) have been neglected for a long time in the study of Alzheimer's disease (AD) pathogenesis. In reference to a new emerging hypothesis according to which there is an altered cerebellar synaptic processing in AD, we verified the possible role played by new biomarkers in the CB of AD patients compared with not-demented healthy control subjects (NDHS). Using a bioinformatics approach, we have collected several microarray datasets and obtained 626 cerebella sample biopsies belonging to subjects who did not die from causes related to neurological diseases and 199 cerebella belonging to AD. The analysis of logical relations between the transcriptome dataset highlighted guanine nucleotide-binding protein (G protein) gamma 13 (GNG13) as a potential new biomarker for Purkinje cells (PCs). We have correlated GNG13 expression levels with already widely existing bibliography of PC marker genes, such as Purkinje cell protein 2 (PCP2), Purkinje cell protein 4 (PCP4), and cerebellin 3 (CBLN3). We showed that expression levels of GNG13 and PCP2, PCP4, and CBLN3 were significantly correlated with each other in NDHS and in AD and significantly reduced in AD patients compared with NDHS subjects. In addition, we highlighted a negative correlation between the expression levels of PC biomarkers and age. From the outcome of our investigation, it is possible to conclude that the identification of GNG13 as a potentially biomarker in PCs represents also a state of health of CB, in association with the expression of PCP2, PCP4, and CBLN3.
Collapse
Affiliation(s)
- Cristina Sanfilippo
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124, Messina, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Maria Kazakova
- Department of Medical Biology, Medical Faculty, Medical University, Plovdiv, Bulgaria
| | - Venera Mazzone
- Department G.F. Ingrassia, Anatomy, School of Medicine, University of Catania, Catania, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
33
|
Brain CHID1 Expression Correlates with NRGN and CALB1 in Healthy Subjects and AD Patients. Cells 2021; 10:cells10040882. [PMID: 33924468 PMCID: PMC8069241 DOI: 10.3390/cells10040882] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease is a progressive, devastating, and irreversible brain disorder that, day by day, destroys memory skills and social behavior. Despite this, the number of known genes suitable for discriminating between AD patients is insufficient. Among the genes potentially involved in the development of AD, there are the chitinase-like proteins (CLPs) CHI3L1, CHI3L2, and CHID1. The genes of the first two have been extensively investigated while, on the contrary, little information is available on CHID1. In this manuscript, we conducted transcriptome meta-analysis on an extensive sample of brains of healthy control subjects (n = 1849) (NDHC) and brains of AD patients (n = 1170) in order to demonstrate CHID1 involvement. Our analysis revealed an inverse correlation between the brain CHID1 expression levels and the age of NDHC subjects. Significant differences were highlighted comparing CHID1 expression of NDHC subjects and AD patients. Exclusive in AD patients, the CHID1 expression levels were correlated positively to calcium-binding adapter molecule 1 (IBA1) levels. Furthermore, both in NDHC and in AD patient’s brains, the CHID1 expression levels were directly correlated with calbindin 1 (CALB1) and neurogranin (NRGN). According to brain regions, correlation differences were shown between the expression levels of CHID1 in prefrontal, frontal, occipital, cerebellum, temporal, and limbic system. Sex-related differences were only highlighted in NDHC. CHID1 represents a new chitinase potentially involved in the principal processes underlying Alzheimer’s disease.
Collapse
|
34
|
Gaudreault R, Hervé V, van de Ven TGM, Mousseau N, Ramassamy C. Polyphenol-Peptide Interactions in Mitigation of Alzheimer's Disease: Role of Biosurface-Induced Aggregation. J Alzheimers Dis 2021; 81:33-55. [PMID: 33749653 DOI: 10.3233/jad-201549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder, responsible for nearly two-thirds of all dementia cases. In this review, we report the potential AD treatment strategies focusing on natural polyphenol molecules (green chemistry) and more specifically on the inhibition of polyphenol-induced amyloid aggregation/disaggregation pathways: in bulk and on biosurfaces. We discuss how these pathways can potentially alter the structure at the early stages of AD, hence delaying the aggregation of amyloid-β (Aβ) and tau. We also discuss multidisciplinary approaches, combining experimental and modelling methods, that can better characterize the biochemical and biophysical interactions between proteins and phenolic ligands. In addition to the surface-induced aggregation, which can occur on surfaces where protein can interact with other proteins and polyphenols, we suggest a new concept referred as "confinement stability". Here, on the contrary, the adsorption of Aβ and tau on biosurfaces other than Aβ- and tau-fibrils, e.g., red blood cells, can lead to confinement stability that minimizes the aggregation of Aβ and tau. Overall, these mechanisms may participate directly or indirectly in mitigating neurodegenerative diseases, by preventing protein self-association, slowing down the aggregation processes, and delaying the progression of AD.
Collapse
Affiliation(s)
- Roger Gaudreault
- Department of Physics, Université de Montréal, Montreal, QC, Canada
| | - Vincent Hervé
- INRS-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | | | - Normand Mousseau
- Department of Physics, Université de Montréal, Montreal, QC, Canada
| | | |
Collapse
|
35
|
Ahmed R, Melacini G. A biophysical toolset to probe the microscopic processes underlying protein aggregation and its inhibition by molecular chaperones. Biophys Chem 2021; 269:106508. [PMID: 33310607 DOI: 10.1016/j.bpc.2020.106508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/13/2020] [Indexed: 10/22/2022]
Abstract
Given the breadth and depth of the scientific contributions of Sir Christopher Dobson, with over 870 publications to date, it is inconceivable to convey in a single review the impact of his work and its legacy. This review therefore primarily focuses on his contributions to the development of strategies for preventing aberrant protein misfolding. The first section of this review highlights his seminal work on the elucidation of the microscopic nucleation processes underlying protein aggregation. Next, we discuss the specific inhibition of these steps by candidate drugs and biologics, with a particular emphasis on the role of molecular chaperones. In the final section, we review how protein aggregation principles can be exploited for the rational design of novel and more potent aggregation inhibitors. These milestones serve as excellent examples of the profound impact of Dobson's seminal work on fundamental science and its translation into drug discovery.
Collapse
Affiliation(s)
- Rashik Ahmed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4M1, Canada
| | - Giuseppe Melacini
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4M1, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada.
| |
Collapse
|
36
|
Grigolato F, Arosio P. The role of surfaces on amyloid formation. Biophys Chem 2021; 270:106533. [PMID: 33529995 DOI: 10.1016/j.bpc.2020.106533] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 01/02/2023]
Abstract
Interfaces can strongly accelerate or inhibit protein aggregation, destabilizing proteins that are stable in solution or, conversely, stabilizing proteins that are aggregation-prone. Although this behaviour is well-known, our understanding of the molecular mechanisms underlying surface-induced protein aggregation is still largely incomplete. A major challenge is represented by the high number of physico-chemical parameters involved, which are highly specific to the considered combination of protein, surface properties, and solution conditions. The key aspect determining the role of interfaces is the relative propensity of the protein to aggregate at the surface with respect to bulk. In this review, we discuss the multiple molecular determinants that regulate this balance. We summarize current experimental techniques aimed at characterizing protein aggregation at interfaces, and highlight the need to complement experimental analysis with theoretical modelling. In particular, we illustrate how chemical kinetic analysis can be combined with experimental methods to provide insights into the molecular mechanisms underlying surface-induced protein aggregation, under both stagnant and agitation conditions. We summarize recent progress in the study of important amyloids systems, focusing on selected relevant interfaces.
Collapse
Affiliation(s)
- Fulvio Grigolato
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich 8093, Switzerland
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zurich, Zurich 8093, Switzerland.
| |
Collapse
|
37
|
Gracia P, Camino JD, Volpicelli-Daley L, Cremades N. Multiplicity of α-Synuclein Aggregated Species and Their Possible Roles in Disease. Int J Mol Sci 2020; 21:E8043. [PMID: 33126694 PMCID: PMC7663424 DOI: 10.3390/ijms21218043] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
α-Synuclein amyloid aggregation is a defining molecular feature of Parkinson's disease, Lewy body dementia, and multiple system atrophy, but can also be found in other neurodegenerative disorders such as Alzheimer's disease. The process of α-synuclein aggregation can be initiated through alternative nucleation mechanisms and dominated by different secondary processes giving rise to multiple amyloid polymorphs and intermediate species. Some aggregated species have more inherent abilities to induce cellular stress and toxicity, while others seem to be more potent in propagating neurodegeneration. The preference for particular types of polymorphs depends on the solution conditions and the cellular microenvironment that the protein encounters, which is likely related to the distinct cellular locations of α-synuclein inclusions in different synucleinopathies, and the existence of disease-specific amyloid polymorphs. In this review, we discuss our current understanding on the nature and structure of the various types of α-synuclein aggregated species and their possible roles in pathology. Precisely defining these distinct α-synuclein species will contribute to understanding the molecular origins of these disorders, developing accurate diagnoses, and designing effective therapeutic interventions for these highly debilitating neurodegenerative diseases.
Collapse
Affiliation(s)
- Pablo Gracia
- Joint Unit BIFI-IQFR (CSIC), Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, 50018 Zaragoza, Spain; (P.G.); (J.D.C.)
| | - José D. Camino
- Joint Unit BIFI-IQFR (CSIC), Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, 50018 Zaragoza, Spain; (P.G.); (J.D.C.)
| | - Laura Volpicelli-Daley
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Nunilo Cremades
- Joint Unit BIFI-IQFR (CSIC), Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, 50018 Zaragoza, Spain; (P.G.); (J.D.C.)
| |
Collapse
|
38
|
Thompson TB, Chaggar P, Kuhl E, Goriely A, for the Alzheimer’s Disease Neuroimaging Initiative. Protein-protein interactions in neurodegenerative diseases: A conspiracy theory. PLoS Comput Biol 2020; 16:e1008267. [PMID: 33048932 PMCID: PMC7584458 DOI: 10.1371/journal.pcbi.1008267] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 10/23/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases such as Alzheimer's or Parkinson's are associated with the prion-like propagation and aggregation of toxic proteins. A long standing hypothesis that amyloid-beta drives Alzheimer's disease has proven the subject of contemporary controversy; leading to new research in both the role of tau protein and its interaction with amyloid-beta. Conversely, recent work in mathematical modeling has demonstrated the relevance of nonlinear reaction-diffusion type equations to capture essential features of the disease. Such approaches have been further simplified, to network-based models, and offer researchers a powerful set of computationally tractable tools with which to investigate neurodegenerative disease dynamics. Here, we propose a novel, coupled network-based model for a two-protein system that includes an enzymatic interaction term alongside a simple model of aggregate transneuronal damage. We apply this theoretical model to test the possible interactions between tau proteins and amyloid-beta and study the resulting coupled behavior between toxic protein clearance and proteopathic phenomenology. Our analysis reveals ways in which amyloid-beta and tau proteins may conspire with each other to enhance the nucleation and propagation of different diseases, thus shedding new light on the importance of protein clearance and protein interaction mechanisms in prion-like models of neurodegenerative disease.
Collapse
Affiliation(s)
| | - Pavanjit Chaggar
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Ellen Kuhl
- Living Matter Laboratory, Stanford University, Stanford, California, USA
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
39
|
The role of fibril structure and surface hydrophobicity in secondary nucleation of amyloid fibrils. Proc Natl Acad Sci U S A 2020; 117:25272-25283. [PMID: 33004626 PMCID: PMC7568274 DOI: 10.1073/pnas.2002956117] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Alzheimer’s disease affects a rapidly growing number of individuals worldwide. Key unresolved questions relate to the onset and propagation of the disease, linked to the self-assembly of amyloid β peptide into fibrillar and smaller aggregates. This study investigates the propagation of aggregates of amyloid β peptide and asks whether hydrophobic molecular features observed on the fibril surface correlate with its ability to catalyze the formation of new aggregates. This question is motivated by the associated formation of intermediate forms that are toxic to neuronal cells. The results imply that surface catalysis is independent of surface details but requires that the monomers that form the new aggregate can adopt the structure of the parent aggregate without steric clashes. Crystals, nanoparticles, and fibrils catalyze the generation of new aggregates on their surface from the same type of monomeric building blocks as the parent assemblies. This secondary nucleation process can be many orders of magnitude faster than primary nucleation. In the case of amyloid fibrils associated with Alzheimer’s disease, this process leads to the multiplication and propagation of aggregates, whereby short-lived oligomeric intermediates cause neurotoxicity. Understanding the catalytic activity is a fundamental goal in elucidating the molecular mechanisms of Alzheimer’s and associated diseases. Here we explore the role of fibril structure and hydrophobicity by asking whether the V18, A21, V40, and A42 side chains which are exposed on the Aβ42 fibril surface as continuous hydrophobic patches play a role in secondary nucleation. Single, double, and quadruple serine substitutions were made. Kinetic analyses of aggregation data at multiple monomer concentrations reveal that all seven mutants retain the dominance of secondary nucleation as the main mechanism of fibril proliferation. This finding highlights the generality of secondary nucleation and its independence of the detailed molecular structure. Cryo-electron micrographs reveal that the V18S substitution causes fibrils to adopt a distinct morphology with longer twist distance than variants lacking this substitution. Self- and cross-seeding data show that surface catalysis is only efficient between peptides of identical morphology, indicating a templating role of secondary nucleation with structural conversion at the fibril surface. Our findings thus provide clear evidence that the propagation of amyloid fibril strains is possible even in systems dominated by secondary nucleation rather than fragmentation.
Collapse
|
40
|
Kinetic fingerprints differentiate the mechanisms of action of anti-Aβ antibodies. Nat Struct Mol Biol 2020; 27:1125-1133. [PMID: 32989305 DOI: 10.1038/s41594-020-0505-6] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 08/13/2020] [Indexed: 12/28/2022]
Abstract
The amyloid cascade hypothesis, according to which the self-assembly of amyloid-β peptide (Aβ) is a causative process in Alzheimer's disease, has driven many therapeutic efforts for the past 20 years. Failures of clinical trials investigating Aβ-targeted therapies have been interpreted as evidence against this hypothesis, irrespective of the characteristics and mechanisms of action of the therapeutic agents, which are highly challenging to assess. Here, we combine kinetic analyses with quantitative binding measurements to address the mechanism of action of four clinical stage anti-Aβ antibodies, aducanumab, gantenerumab, bapineuzumab and solanezumab. We quantify the influence of these antibodies on the aggregation kinetics and on the production of oligomeric aggregates and link these effects to the affinity and stoichiometry of each antibody for monomeric and fibrillar forms of Aβ. Our results reveal that, uniquely among these four antibodies, aducanumab dramatically reduces the flux of Aβ oligomers.
Collapse
|
41
|
Sanfilippo C, Castrogiovanni P, Imbesi R, Nunnari G, Di Rosa M. Postsynaptic damage and microglial activation in AD patients could be linked CXCR4/CXCL12 expression levels. Brain Res 2020; 1749:147127. [PMID: 32949560 DOI: 10.1016/j.brainres.2020.147127] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/26/2020] [Accepted: 09/12/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is one of the most common forms of dementia with still unknown pathogenesis. Several cytokines and chemokines are involved in the pathogenesis of AD. Among the chemokines, the CXCR4/CXCL12 complex has been shown to play an important role in the pathogenetic development of AD. We investigated the expression levels of CXCR4 / CXCL12 in fifteen brain regions of healthy non-demented subjects (NDHC) (2139 sample) and AD patients (1170 sample) stratified according to sex and age. Furthermore, we correlated their expressions with the Neurogranin (NRGN) and CHI3L1 levels, two inflamm-aging markers. We highlighted that CXCR4 gene expression levels were age-correlated in the brain of NDHC subjects and that AD nullified this correlation. A similar trend, but diametrically opposite was observed for CXCL12. Its expression was decreased during the aging in both sexes, and in the brains of AD patients, it underwent an inversion of the trend, only and exclusively in females. Brains of AD patients expressed high CXCR4 and CHI3L1, and low CXCL12 and Neurogranin levels compared to NDHC subjects. Both CXCR4 and CXCL12 correlated significantly with CHI3L1 and Neurogranin expression levels, regardless of disease. Furthermore, we showed a selective modulation of CXCL12 and CXCR4 only in specific brain regions. Taken together our results demonstrate that CXCL12 and CXCR4 are linked to Neurogranin and CHI3L1 expression levels and the relationship between postsynaptic damage and microglial activation in AD could be shown using all these genes. Further confirmations are needed to demonstrate the close link between these genes.
Collapse
Affiliation(s)
- Cristina Sanfilippo
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Italy
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Italy
| | - Giuseppe Nunnari
- Unit of Infectious Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Italy.
| |
Collapse
|
42
|
Chen X, Dong GY, Wang LX. High-frequency transcranial magnetic stimulation protects APP/PS1 mice against Alzheimer's disease progress by reducing APOE and enhancing autophagy. Brain Behav 2020; 10:e01740. [PMID: 32592331 PMCID: PMC7428505 DOI: 10.1002/brb3.1740] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/16/2020] [Accepted: 06/07/2020] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION The repetitive transcranial magnetic stimulation (rTMS) has clinically wide application prospect of psychiatry and neuroscience, for its painless, noninvasive, and high efficiency. So far, rTMS has been used in the treatment of Alzheimer's disease (AD) but the underlying mechanism is not clear. METHODS AND RESULTS The APP/PS1 mice at 3-month-old were treated by 5 Hz high-frequency (HF) rTMS for two weeks. After rTMS treatment, the AD-like cognitive impairments of APP/PS1 mice were investigated subsequently, and molecular mechanisms underlying was further explored. The study showed that the 2-week rTMS at 5Hz frequency improved cognitive impairments and AD-like pathology (including a decrease in p-Tau, APP, Aβ, and PP2A expression) of APP/PS1 mice. Although BDNF-TrkB signaling was significantly enhanced, no differences of SYN, PSD95 and p-AKT were observed in the brain of APP/PS1 mice. On the contrary, the LC3Ⅱ/LC3Ⅰ ratio was elevated with a significant reduction of ApoE and p62 in mice. CONCLUSIONS rTMS exerts a potentially protective role in the prevention and treatment of AD by reducing ApoE expression and promoting autophagic flux, which provides a new insight into the mechanism of rTMS.
Collapse
Affiliation(s)
- Xia Chen
- Department of Neurology, Laboratory of Neurological Diseases, Reproductive Medicine Centre, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Guo-Ying Dong
- Department of Neurology, Laboratory of Neurological Diseases, Reproductive Medicine Centre, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Lin-Xiao Wang
- Department of Neurology, Laboratory of Neurological Diseases, Reproductive Medicine Centre, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
43
|
CHI3L2 Expression Levels Are Correlated with AIF1, PECAM1, and CALB1 in the Brains of Alzheimer's Disease Patients. J Mol Neurosci 2020; 70:1598-1610. [PMID: 32705525 DOI: 10.1007/s12031-020-01667-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) represents one of the main forms of dementia that afflicts our society. The expression of several genes has been associated with disease development. Despite this, the number of genes known to be capable of discriminating between AD patients according to sex remains deficient. In our study, we performed a transcriptomes meta-analysis on a large court of brains of healthy control subjects (n = 2139) (NDHC) and brains of AD patients (n = 1170). Our aim was to verify the brain expression levels of CHI3L2 and its correlation with genes associated with microglia-mediated neuroinflammation (IBA1), alteration of the blood-brain barrier (PECAM1), and neuronal damage (CALB1). We showed that the CHI3L2, IBA1, PECAM1, and CALB1 expression levels were modulated in the brains of patients with AD compared to NDHC subjects. Furthermore, both in NDHC and in AD patient's brains, the CHI3L2 expression levels were directly correlated with IBA1 and PECAM1 and inversely with CALB1. Additionally, the expression levels of CHI3L2, PECAM1, and CALB1 but not of IBA1 were sex-depended. By stratifying the samples according to age and sex, correlation differences emerged between the expression levels of CHI3L2, IBA1, PECAM1, and CALB1 and the age of NDHC subjects and AD patients. CHI3L2 represents a promising gene potentially involved in the key processes underlying Alzheimer's disease. Its expression in the brains of sex-conditioned AD patients opens up new possible sex therapeutic strategies aimed at controlling imbalance in disease progression.
Collapse
|
44
|
Robinson CV. Christopher Dobson, 1949-2019: Mentor, Friend, Scientist Extraordinaire. Annu Rev Biochem 2020; 89:1-19. [PMID: 32343910 DOI: 10.1146/annurev-biochem-011520-105226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
It is impossible to do justice in one review article to a researcher of the stature of Christopher Dobson. His career spanned almost five decades, resulting in more than 870 publications and a legacy that will continue to influence the lives of many for decades to come. In this review, I have attempted to capture Chris's major contributions: his early work, dedicated to understanding protein-folding mechanisms; his collaborative work with physicists to understand the process of protein aggregation; and finally, his later career in which he developed strategies to prevent misfolding. However, it is not only this body of work but also the man himself who inspired an entire generation of scientists through his patience, ability to mentor, and innate generosity. These qualities remain a hallmark of the way in which he conducted his research-research that will leave a lasting imprint on science.
Collapse
Affiliation(s)
- Carol V Robinson
- Department of Physical and Theoretical Chemistry, University of Oxford, Oxford OX1 3QZ, United Kingdom;
| |
Collapse
|
45
|
Ultrastructural evidence for self-replication of Alzheimer-associated Aβ42 amyloid along the sides of fibrils. Proc Natl Acad Sci U S A 2020; 117:11265-11273. [PMID: 32439711 PMCID: PMC7260961 DOI: 10.1073/pnas.1918481117] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Two unresolved problems in Alzheimer’s disease (AD) are its onset and propagation, linked to Aβ peptide aggregation. Fibrils of Aβ42 may grow by monomer addition at their ends. Additionally, through so-called secondary nucleation, fibrils can catalyse the formation of new aggregates from monomer on their surface, thereby generating oligomeric species that are toxic to brain tissue. Insights into the structural transitions occurring during secondary nucleation will facilitate the design of therapies to limit the neurotoxicity in AD, but such information is currently lacking. This study identifies conditions that allow the capture of reaction intermediates of secondary nucleation for the purpose of ultrastructural characterization. These reaction intermediates are morphologically distinct from mature fibrils and cover the sides of fibrils during an on-going aggregation reaction. The nucleation of Alzheimer-associated Aβ peptide monomers can be catalyzed by preexisting Aβ fibrils. This leads to autocatalytic amplification of aggregate mass and underlies self-replication and generation of toxic oligomers associated with several neurodegenerative diseases. However, the nature of the interactions between the monomeric species and the fibrils during this key process, and indeed the ultrastructural localization of the interaction sites have remained elusive. Here we used NMR and optical spectroscopy to identify conditions that enable the capture of transient species during the aggregation and secondary nucleation of the Aβ42 peptide. Cryo-electron microscopy (cryo-EM) images show that new aggregates protrude from the entire length of the progenitor fibril. These protrusions are morphologically distinct from the well-ordered fibrils dominating at the end of the aggregation process. The data provide direct evidence that self-replication through secondary nucleation occurs along the sides of fibrils, which become heavily decorated under the current solution conditions (14 µM Aβ42, 20 mM sodium phosphate, 200 µM EDTA, pH 6.8).
Collapse
|
46
|
Fornari S, Schäfer A, Kuhl E, Goriely A. Spatially-extended nucleation-aggregation-fragmentation models for the dynamics of prion-like neurodegenerative protein-spreading in the brain and its connectome. J Theor Biol 2019; 486:110102. [PMID: 31809717 DOI: 10.1016/j.jtbi.2019.110102] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/30/2019] [Accepted: 11/29/2019] [Indexed: 12/20/2022]
Abstract
The prion-like hypothesis of neurodegenerative diseases states that the accumulation of misfolded proteins in the form of aggregates is responsible for tissue death and its associated neurodegenerative pathology and cognitive decline. Some disease-specific misfolded proteins can interact with healthy proteins to form long chains that are transported through the brain along axonal pathways. Since aggregates of different sizes have different transport properties and toxicity, it is important to follow independently their evolution in space and time. Here, we model the spreading and propagation of aggregates of misfolded proteins in the brain using the general Smoluchowski theory of nucleation, aggregation, and fragmentation. The transport processes considered here are either anisotropic diffusion along axonal bundles or discrete Laplacian transport along a network. In particular, we model the spreading and aggregation of both amyloid-β and τ molecules in the brain connectome. We show that these two models lead to different size distributions and different propagation along the network. A detailed analysis of these two models also reveals the existence of four different stages with different dynamics and invasive properties.
Collapse
Affiliation(s)
- Sveva Fornari
- Living Matter Laboratory, Stanford University, Stanford, USA
| | - Amelie Schäfer
- Living Matter Laboratory, Stanford University, Stanford, USA
| | - Ellen Kuhl
- Living Matter Laboratory, Stanford University, Stanford, USA
| | - Alain Goriely
- Mathematical Institute, University of Oxford, Oxford, UK.
| |
Collapse
|