1
|
Vargas-Rosales PA, Caflisch A. The physics-AI dialogue in drug design. RSC Med Chem 2025; 16:1499-1515. [PMID: 39906313 PMCID: PMC11788922 DOI: 10.1039/d4md00869c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
A long path has led from the determination of the first protein structure in 1960 to the recent breakthroughs in protein science. Protein structure prediction and design methodologies based on machine learning (ML) have been recognized with the 2024 Nobel prize in Chemistry, but they would not have been possible without previous work and the input of many domain scientists. Challenges remain in the application of ML tools for the prediction of structural ensembles and their usage within the software pipelines for structure determination by crystallography or cryogenic electron microscopy. In the drug discovery workflow, ML techniques are being used in diverse areas such as scoring of docked poses, or the generation of molecular descriptors. As the ML techniques become more widespread, novel applications emerge which can profit from the large amounts of data available. Nevertheless, it is essential to balance the potential advantages against the environmental costs of ML deployment to decide if and when it is best to apply it. For hit to lead optimization ML tools can efficiently interpolate between compounds in large chemical series but free energy calculations by molecular dynamics simulations seem to be superior for designing novel derivatives. Importantly, the potential complementarity and/or synergism of physics-based methods (e.g., force field-based simulation models) and data-hungry ML techniques is growing strongly. Current ML methods have evolved from decades of research. It is now necessary for biologists, physicists, and computer scientists to fully understand advantages and limitations of ML techniques to ensure that the complementarity of physics-based methods and ML tools can be fully exploited for drug design.
Collapse
Affiliation(s)
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich Winterthurerstrasse 190 8057 Zürich Switzerland
| |
Collapse
|
2
|
Westermark GT, Nyström E, Nyström S, Nilsson KPR, Hammarström P, Westermark P. The question of strains in AA amyloidosis. Sci Rep 2025; 15:3684. [PMID: 39881136 PMCID: PMC11779915 DOI: 10.1038/s41598-025-87239-6] [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: 10/14/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
The existence of transmissible amyloid fibril strains has long intrigued the scientific community. The strain theory originates from prion disorders, but here, we provide evidence of strains in systemic amyloidosis. Human AA amyloidosis manifests as two distinct clinical phenotypes called common AA and vascular AA. Glomerular amyloid deposition of the kidney defines the common form, while in the vascular type amyloid deposits are massive in the renal medulla and in arteries throughout the body, while glomeruli are spared. By electron microscopy the two types appeared morphologically different. The common type was composed of dispersed fibrils which tended to be clustered whereas the vascular type was composed of longer and more distinct less clustered fibrils. Staining with fluorescent amyloid binding ligands analyzed by hyperspectral microscopy showed differential staining patterns between the two groups supporting the notion of human AA amyloid strains. AA amyloid staining was significantly different from systemic AL amyloid. Both types of AA (common and vascular) and AL amyloid fibrils were isolated and used to seed mouse AA amyloid in groups of inflamed NMRI mice (n = 9-10 per group). All but two mice showed amyloid deposits in the spleen induced by the human seeds. Amyloid binding ligand analysis was applied on the splenic amyloid deposits and revealed no clear significant difference between mice seeded with AA fibrils from different donors being vascular or common, but the AA deposits of mice given AL fibrils showed significantly different amyloid fluorescent signals compared to all groups of mice receiving AA fibrils. The combined results support the hypothesis that AA amyloid fibril structures can vary depending on the seed and may manifest as amyloid strains.
Collapse
Affiliation(s)
| | - Ebba Nyström
- Department of Physics, Chemistry and Biology (IFM), Linköping University, 58183, Linköping, Sweden
| | - Sofie Nyström
- Department of Physics, Chemistry and Biology (IFM), Linköping University, 58183, Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology (IFM), Linköping University, 58183, Linköping, Sweden
| | - Per Hammarström
- Department of Physics, Chemistry and Biology (IFM), Linköping University, 58183, Linköping, Sweden
| | - Per Westermark
- Department of Immunology, Genetics and Pathology, Uppsala University, Rudbeck Laboratory, C11, 75185, Uppsala, Sweden.
| |
Collapse
|
3
|
Klingstedt T, Shirani H, Parvin F, Nyström S, Hammarström P, Graff C, Ingelsson M, Vidal R, Ghetti B, Sehlin D, Syvänen S, Nilsson KPR. Dual-ligand fluorescence microscopy enables chronological and spatial histological assignment of distinct amyloid-β deposits. J Biol Chem 2025; 301:108032. [PMID: 39615691 PMCID: PMC11731580 DOI: 10.1016/j.jbc.2024.108032] [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: 08/14/2024] [Revised: 10/25/2024] [Accepted: 11/21/2024] [Indexed: 12/23/2024] Open
Abstract
Different types of deposits comprised of amyloid-β (Aβ) peptides are one of the pathological hallmarks of Alzheimer's disease (AD) and novel methods that enable identification of a diversity of Aβ deposits during the AD continuum are essential for understanding the role of these aggregates during the pathogenesis. Herein, different combinations of five fluorescent thiophene-based ligands were used for detection of Aβ deposits in brain tissue sections from transgenic mouse models with aggregated Aβ pathology, as well as brain tissue sections from patients affected by sporadic or dominantly inherited AD. When analyzing the sections with fluorescence microscopy, distinct ligand staining patterns related to the transgenic mouse model or to the age of the mice were observed. Likewise, specific staining patterns of different Aβ deposits were revealed for sporadic versus dominantly inherited AD, as well as for distinct brain regions in sporadic AD. Thus, by using dual-staining protocols with multiple combinations of fluorescent ligands, a chronological and spatial histological designation of different Aβ deposits could be achieved. This study demonstrates the potential of our approach for resolving the role and presence of distinct Aβ aggregates during the AD continuum and pinpoints the necessity of using multiple ligands to obtain an accurate assignment of different Aβ deposits in the neuropathological evaluation of AD, as well as when evaluating therapeutic strategies targeting Aβ aggregates.
Collapse
Affiliation(s)
- Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Hamid Shirani
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Farjana Parvin
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Sofie Nyström
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Per Hammarström
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Caroline Graff
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Martin Ingelsson
- Krembil Brain Institute, University Health Network, Toronto, Ontario, Canada; Tanz Centre for Research in Neurodegenerative Diseases, Department of Medicine and Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada; Molecular Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Ruben Vidal
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dag Sehlin
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Stina Syvänen
- Molecular Geriatrics, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden.
| |
Collapse
|
4
|
Swaminathan P, Klingstedt T, Theologidis V, Gram H, Larsson J, Hagen L, Liabakk NB, Gederaas OA, Hammarström P, Nilsson KPR, Van Den Berge N, Lindgren M. In Vitro Cell Model Investigation of Alpha-Synuclein Aggregate Morphology Using Spectroscopic Imaging. Int J Mol Sci 2024; 25:12458. [PMID: 39596523 PMCID: PMC11594916 DOI: 10.3390/ijms252212458] [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: 10/25/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Recently, it has been hypothesized that alpha-synuclein protein strain morphology may be associated with clinical subtypes of alpha-synucleinopathies, like Parkinson's disease and multiple system atrophy. However, direct evidence is lacking due to the caveat of conformation-specific characterization of protein strain morphology. Here we present a new cell model based in vitro method to explore various alpha-synuclein (αsyn) aggregate morphotypes. We performed a spectroscopic investigation of the HEK293 cell model, transfected with human wildtype-αsyn and A53T-αsyn variants, using the amyloid fibril-specific heptameric luminescent oligomeric thiophene h-FTAA. The spectral profile of h-FTAA binding to aggregates displayed a blue-shifted spectrum with a fluorescence decay time longer than in PBS, suggesting a hydrophobic binding site. In vitro spectroscopic binding characterization of h-FTAA with αsyn pre-formed fibrils suggested a binding dissociation constant Kd < 100 nM. The cells expressing the A53T-αsyn and human wildtype-αsyn were exposed to recombinant pre-formed fibrils of human αsyn. The ensuing intracellular aggregates were stained with h-FTAA followed by an evaluation of the spectral features and fluorescence lifetime of intracellular αsyn/h-FTAA, in order to characterize aggregate morphotypes. This study exemplifies the use of cell culture together with conformation-specific ligands to characterize strain morphology by investigating the spectral profiles and fluorescence lifetime of h-FTAA, based upon its binding to a certain αsyn aggregate. This study paves the way for toxicity studies of different αsyn strains in vitro and in vivo. Accurate differentiation of specific alpha-synucleinopathies is still limited to advanced disease stages. However, early subtype-specific diagnosis is of the utmost importance for prognosis and treatment response. The potential association of αsyn aggregates morphotypes detected in biopsies or fluids to disease phenotypes would allow for subtype-specific diagnosis in subclinical disease stage and potentially reveal new subtype-specific treatment targets. Notably, the method may be applied to the entire spectrum of neurodegenerative diseases by using a combination of conformation-specific ligands in a physicochemical environment together with other types of polymorphic amyloid variants and assess the conformation-specific features of various protein pathologies.
Collapse
Affiliation(s)
- Priyanka Swaminathan
- Department of Physics, Faculty of Natural Sciences, Norwegian University of Science and Technology (NTNU), Gløshaugen, Realfagbygget, NO-7491 Trondheim, Norway;
| | - Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - Vasileios Theologidis
- Department of Clinical Medicine—Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, 8000 Aarhus, Denmark
| | - Hjalte Gram
- DANDRITE, Danish Research Institute of Translational Neuroscience & Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Johan Larsson
- Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - Lars Hagen
- PROMEC—Proteomics and Modomics Experimental Core Facility at NTNU and the Central Norway Regional Health Authority, NO-7491 Trondheim, Norway
| | - Nina B. Liabakk
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Odrun A. Gederaas
- Division of Natural Sciences, University of Agder, NO-4630 Kristiansand, Norway
| | - Per Hammarström
- Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - K. Peter R. Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, SE-581 83 Linköping, Sweden
| | - Nathalie Van Den Berge
- Department of Clinical Medicine—Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus University, 8000 Aarhus, Denmark
| | - Mikael Lindgren
- Department of Physics, Faculty of Natural Sciences, Norwegian University of Science and Technology (NTNU), Gløshaugen, Realfagbygget, NO-7491 Trondheim, Norway;
| |
Collapse
|
5
|
Sobek J, Li J, Combes BF, Gerez JA, Henrich MT, Geibl FF, Nilsson PR, Shi K, Rominger A, Oertel WH, Nitsch RM, Nordberg A, Ågren H, Ni R. Efficient characterization of multiple binding sites of small molecule imaging ligands on amyloid-beta, tau and alpha-synuclein. Eur J Nucl Med Mol Imaging 2024; 51:3960-3977. [PMID: 38953933 PMCID: PMC11527973 DOI: 10.1007/s00259-024-06806-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE There is an unmet need for compounds to detect fibrillar forms of alpha-synuclein (αSyn) and 4-repeat tau, which are critical in many neurodegenerative diseases. Here, we aim to develop an efficient surface plasmon resonance (SPR)-based assay to facilitate the characterization of small molecules that can bind these fibrils. METHODS SPR measurements were conducted to characterize the binding properties of fluorescent ligands/compounds toward recombinant amyloid-beta (Aβ)42, K18-tau, full-length 2N4R-tau and αSyn fibrils. In silico modeling was performed to examine the binding pockets of ligands on αSyn fibrils. Immunofluorescence staining of postmortem brain tissue slices from Parkinson's disease patients and mouse models was performed with fluorescence ligands and specific antibodies. RESULTS We optimized the protocol for the immobilization of Aβ42, K18-tau, full-length 2N4R-tau and αSyn fibrils in a controlled aggregation state on SPR-sensor chips and for assessing their binding to ligands. The SPR results from the analysis of binding kinetics suggested the presence of at least two binding sites for all fibrils, including luminescent conjugated oligothiophenes, benzothiazole derivatives, nonfluorescent methylene blue and lansoprazole. In silico modeling studies for αSyn (6H6B) revealed four binding sites with a preference for one site on the surface. Immunofluorescence staining validated the detection of pS129-αSyn positivity in the brains of Parkinson's disease patients and αSyn preformed-fibril injected mice, 6E10-positive Aβ in arcAβ mice, and AT-8/AT-100-positivity in pR5 mice. CONCLUSION SPR measurements of small molecules binding to Aβ42, K18/full-length 2N4R-tau and αSyn fibrils suggested the existence of multiple binding sites. This approach may provide efficient characterization of compounds for neurodegenerative disease-relevant proteinopathies.
Collapse
Affiliation(s)
- Jens Sobek
- Functional Genomics Center, University of Zurich & ETH Zurich, Zürich, Switzerland
| | - Junhao Li
- Department of Physics and Astronomy, Uppsala University, Uppsala, Sweden
| | - Benjamin F Combes
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Zürich, Switzerland
| | - Juan A Gerez
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Zürich, Switzerland
| | - Martin T Henrich
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - Fanni F Geibl
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - Peter R Nilsson
- Divison of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Wolfgang H Oertel
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Zürich, Switzerland
| | - Agneta Nordberg
- Divison of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hans Ågren
- Department of Physics and Astronomy, Uppsala University, Uppsala, Sweden
| | - Ruiqing Ni
- Institute for Regenerative Medicine, University of Zurich, Wagistrasse 12, 8952, Zürich, Switzerland.
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zürich, Switzerland.
| |
Collapse
|
6
|
de Raffele D, Ilie IM. Unlocking novel therapies: cyclic peptide design for amyloidogenic targets through synergies of experiments, simulations, and machine learning. Chem Commun (Camb) 2024; 60:632-645. [PMID: 38131333 DOI: 10.1039/d3cc04630c] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Existing therapies for neurodegenerative diseases like Parkinson's and Alzheimer's address only their symptoms and do not prevent disease onset. Common therapeutic agents, such as small molecules and antibodies struggle with insufficient selectivity, stability and bioavailability, leading to poor performance in clinical trials. Peptide-based therapeutics are emerging as promising candidates, with successful applications for cardiovascular diseases and cancers due to their high bioavailability, good efficacy and specificity. In particular, cyclic peptides have a long in vivo stability, while maintaining a robust antibody-like binding affinity. However, the de novo design of cyclic peptides is challenging due to the lack of long-lived druggable pockets of the target polypeptide, absence of exhaustive conformational distributions of the target and/or the binder, unknown binding site, methodological limitations, associated constraints (failed trials, time, money) and the vast combinatorial sequence space. Hence, efficient alignment and cooperation between disciplines, and synergies between experiments and simulations complemented by popular techniques like machine-learning can significantly speed up the therapeutic cyclic-peptide development for neurodegenerative diseases. We review the latest advancements in cyclic peptide design against amyloidogenic targets from a computational perspective in light of recent advancements and potential of machine learning to optimize the design process. We discuss the difficulties encountered when designing novel peptide-based inhibitors and we propose new strategies incorporating experiments, simulations and machine learning to design cyclic peptides to inhibit the toxic propagation of amyloidogenic polypeptides. Importantly, these strategies extend beyond the mere design of cyclic peptides and serve as template for the de novo generation of (bio)materials with programmable properties.
Collapse
Affiliation(s)
- Daria de Raffele
- University of Amsterdam, van 't Hoff Institute for Molecular Sciences, Science Park 904, P.O. Box 94157, 1090 GD Amsterdam, The Netherlands.
- Amsterdam Center for Multiscale Modeling (ACMM), University of Amsterdam, P.O. Box 94157, 1090 GD Amsterdam, The Netherlands
| | - Ioana M Ilie
- University of Amsterdam, van 't Hoff Institute for Molecular Sciences, Science Park 904, P.O. Box 94157, 1090 GD Amsterdam, The Netherlands.
- Amsterdam Center for Multiscale Modeling (ACMM), University of Amsterdam, P.O. Box 94157, 1090 GD Amsterdam, The Netherlands
| |
Collapse
|
7
|
Louros N, Schymkowitz J, Rousseau F. Mechanisms and pathology of protein misfolding and aggregation. Nat Rev Mol Cell Biol 2023; 24:912-933. [PMID: 37684425 DOI: 10.1038/s41580-023-00647-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 09/10/2023]
Abstract
Despite advances in machine learning-based protein structure prediction, we are still far from fully understanding how proteins fold into their native conformation. The conventional notion that polypeptides fold spontaneously to their biologically active states has gradually been replaced by our understanding that cellular protein folding often requires context-dependent guidance from molecular chaperones in order to avoid misfolding. Misfolded proteins can aggregate into larger structures, such as amyloid fibrils, which perpetuate the misfolding process, creating a self-reinforcing cascade. A surge in amyloid fibril structures has deepened our comprehension of how a single polypeptide sequence can exhibit multiple amyloid conformations, known as polymorphism. The assembly of these polymorphs is not a random process but is influenced by the specific conditions and tissues in which they originate. This observation suggests that, similar to the folding of native proteins, the kinetics of pathological amyloid assembly are modulated by interactions specific to cells and tissues. Here, we review the current understanding of how intrinsic protein conformational propensities are modulated by physiological and pathological interactions in the cell to shape protein misfolding and aggregation pathology.
Collapse
Affiliation(s)
- Nikolaos Louros
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| | - Frederic Rousseau
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
8
|
Tao Y, Xia W, Zhao Q, Xiang H, Han C, Zhang S, Gu W, Tang W, Li Y, Tan L, Li D, Liu C. Structural mechanism for specific binding of chemical compounds to amyloid fibrils. Nat Chem Biol 2023; 19:1235-1245. [PMID: 37400537 DOI: 10.1038/s41589-023-01370-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/26/2023] [Indexed: 07/05/2023]
Abstract
Amyloid fibril is an important pharmaceutical target for diagnostic and therapeutic treatment of neurodegenerative diseases. However, rational design of chemical compounds that interact with amyloid fibrils is unachievable due to the lack of mechanistic understanding of the ligand-fibril interaction. Here we used cryoelectron microscopy to survey the amyloid fibril-binding mechanism of a series of compounds including classic dyes, (pre)clinical imaging tracers and newly identified binders from high-throughput screening. We obtained clear densities of several compounds in complex with an α-synuclein fibril. These structures unveil the basic mechanism of the ligand-fibril interaction, which exhibits remarkable difference from the canonical ligand-protein interaction. In addition, we discovered a druggable pocket that is also conserved in the ex vivo α-synuclein fibrils from multiple system atrophy. Collectively, these findings expand our knowledge of protein-ligand interaction in the amyloid fibril state, which will enable rational design of amyloid binders in a medicinally beneficial way.
Collapse
Affiliation(s)
- Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Huaijiang Xiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Chao Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Gu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Wenjun Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ying Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Li Tan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- University of the Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
9
|
Vargas-Rosales P, D’Addio A, Zhang Y, Caflisch A. Disrupting Dimeric β-Amyloid by Electric Fields. ACS PHYSICAL CHEMISTRY AU 2023; 3:456-466. [PMID: 37780539 PMCID: PMC10540290 DOI: 10.1021/acsphyschemau.3c00021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 10/03/2023]
Abstract
The early oligomers of the amyloid Aβ peptide are implicated in Alzheimer's disease, but their transient nature complicates the characterization of their structure and toxicity. Here, we investigate the stability of the minimal toxic species, i.e., β-amyloid dimers, in the presence of an oscillating electric field. We first use deep learning (AlphaFold-multimer) for generating initial models of Aβ42 dimers. The flexibility and secondary structure content of the models are then analyzed by multiple runs of molecular dynamics (MD). Structurally stable models are similar to ensemble representatives from microsecond-long MD sampling. Finally, we employ the validated model as the starting structure of MD simulations in the presence of an external oscillating electric field and observe a fast decay of β-sheet content at high field strengths. Control simulations using the helical dimer of the 42-residue leucine zipper peptide show higher structural stability than the Aβ42 dimer. The simulation results provide evidence that an external electric field (oscillating at 1 GHz) can disrupt amyloid oligomers which should be further investigated by experiments with brain organoids in vitro and eventually in vivo.
Collapse
Affiliation(s)
| | - Alessio D’Addio
- Department of Biochemistry, University of Zurich, CH-8057 Zürich, Switzerland
| | - Yang Zhang
- Department of Biochemistry, University of Zurich, CH-8057 Zürich, Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich, CH-8057 Zürich, Switzerland
| |
Collapse
|
10
|
Richter-Dahlfors A, Kärkkäinen E, Choong FX. Fluorescent optotracers for bacterial and biofilm detection and diagnostics. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2023; 24:2246867. [PMID: 37680974 PMCID: PMC10481766 DOI: 10.1080/14686996.2023.2246867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/03/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023]
Abstract
Effective treatment of bacterial infections requires methods that accurately and quickly identify which antibiotic should be prescribed. This review describes recent research on the development of optotracing methodologies for bacterial and biofilm detection and diagnostics. Optotracers are small, chemically well-defined, anionic fluorescent tracer molecules that detect peptide- and carbohydrate-based biopolymers. This class of organic molecules (luminescent conjugated oligothiophenes) show unique electronic, electrochemical and optical properties originating from the conjugated structure of the compounds. The photophysical properties are further improved as donor-acceptor-donor (D-A-D)-type motifs are incorporated in the conjugated backbone. Optotracers bind their biopolymeric target molecules via electrostatic interactions. Binding alters the optical properties of these tracer molecules, shown as altered absorption and emission spectra, as well as ON-like switch of fluorescence. As the optotracer provides a defined spectral signature for each binding partner, a fingerprint is generated that can be used for identification of the target biopolymer. Alongside their use for in situ experimentation, optotracers have demonstrated excellent use in studies of a number of clinically relevant microbial pathogens. These methods will find widespread use across a variety of communities engaged in reducing the effect of antibiotic resistance. This includes basic researchers studying molecular resistance mechanisms, academia and pharma developing new antimicrobials targeting biofilm infections and tests to diagnose biofilm infections, as well as those developing antibiotic susceptibility tests for biofilm infections (biofilm-AST). By iterating between the microbial world and that of plants, development of the optotracing technology has become a prime example of successful cross-feeding across the boundaries of disciplines. As optotracers offers a capacity to redefine the way we work with polysaccharides in the microbial world as well as with plant biomass, the technology is providing novel outputs desperately needed for global impact of the threat of antimicrobial resistance as well as our strive for a circular bioeconomy.
Collapse
Affiliation(s)
- Agneta Richter-Dahlfors
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Fiber and Polymer Technology, School of Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Elina Kärkkäinen
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ferdinand X. Choong
- AIMES – Center for the Advancement of Integrated Medical and Engineering Sciences at Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Björk L, Klingstedt T, Nilsson KPR. Thiophene-Based Ligands: Design, Synthesis and Their Utilization for Optical Assignment of Polymorphic-Disease-Associated Protein Aggregates. Chembiochem 2023; 24:e202300044. [PMID: 36891883 PMCID: PMC10404026 DOI: 10.1002/cbic.202300044] [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: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/10/2023]
Abstract
The development of ligands for detecting protein aggregates is of great interest, as these aggregated proteinaceous species are the pathological hallmarks of several devastating diseases, including Alzheimer's disease. In this regard, thiophene-based ligands have emerged as powerful tools for fluorescent assessment of these pathological entities. The intrinsic conformationally sensitive photophysical properties of poly- and oligothiophenes have allowed optical assignment of disease-associated protein aggregates in tissue sections, as well as real-time in vivo imaging of protein deposits. Herein, we recount the chemical evolution of different generations of thiophene-based ligands, and exemplify their use for the optical distinction of polymorphic protein aggregates. Furthermore, the chemical determinants for achieving a superior fluorescent thiophene-based ligand, as well as the next generation of thiophene-based ligands targeting distinct aggregated species are described. Finally, the directions for future research into the chemical design of thiophene-based ligands that can aid in resolving the scientific challenges around protein aggregation diseases are discussed.
Collapse
Affiliation(s)
- Linnea Björk
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| |
Collapse
|
12
|
Lantz L, Shirani H, Ghetti B, Vidal R, Klingstedt T, Nilsson KPR. Thiophene-Based Ligands for Histological Multiplex Spectral Detection of Distinct Protein Aggregates in Alzheimer's Disease. Chemistry 2023; 29:e202203568. [PMID: 36645413 PMCID: PMC10101888 DOI: 10.1002/chem.202203568] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/17/2023]
Abstract
The aggregation and accumulation of proteins in the brain is the defining feature of many devastating neurodegenerative diseases. The development of fluorescent ligands that bind to these accumulations, or deposits, is essential for the characterization of these neuropathological lesions. We report the synthesis of donor-acceptor-donor (D-A-D) thiophene-based ligands with different emission properties. The D-A-D ligands displayed selectivity towards distinct disease-associated protein deposits in histological sections from postmortem brain tissue of individuals affected by Alzheimer's disease (AD). The ability of the ligands to selectively identify AD-associated pathological alterations, such as deposits composed of aggregates of the amyloid-β (Aβ) peptide or tau, was reduced when the chemical composition of the ligands was altered. When combining the D-A-D ligands with conventional thiophene-based ligands, superior spectral separation of distinct protein aggregates in AD tissue sections was obtained. Our findings provide the structural and functional basis for the development of new fluorescent ligands that can distinguish between aggregated proteinaceous species, as well as offer novel strategies for developing multiplex fluorescence detection of protein aggregates in tissue sections.
Collapse
Affiliation(s)
- Linda Lantz
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Hamid Shirani
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, 46202, Indiana, USA
| | - Ruben Vidal
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, 46202, Indiana, USA
| | - Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| |
Collapse
|
13
|
Sulheim E, WiderØe M, Bäck M, Nilsson KPR, Hammarström P, Nilsson LN, Davies CDL, Åslund AK. Contrast Enhanced Magnetic Resonance Imaging of Amyloid-β Plaques in a Murine Alzheimer’s Disease Model. J Alzheimers Dis 2023; 93:411-419. [PMID: 37038807 DOI: 10.3233/jad-220198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Background: Early detection of amyloid-β (Aβ) aggregates is a critical step to improve the treatment of Alzheimer’s disease (AD) because neuronal damage by the Aβ aggregates occurs before clinical symptoms are apparent. We have previously shown that luminescent conjugated oligothiophenes (LCOs), which are highly specific towards protein aggregates of Aβ, can be used to fluorescently label amyloid plaque in living rodents. Objective: We hypothesize that the LCO can be used to target gadolinium to the amyloid plaque and hence make the plaque detectable by T1-weighted magnetic resonance imaging (MRI). Methods: A novel LCO-gadolinium construct was synthesized to selectively bind to Aβ plaques and give contrast in conventional T1-weighted MR images after intravenous injection in Tg-APPSwe mice. Results: We found that mice with high plaque-burden could be identified using the LCO-Gd constructs by conventional MRI. Conclusion: Our study shows that MR imaging of amyloid plaques is challenging but feasible, and hence contrast-mediated MR imaging could be a valuable tool for early AD detection.
Collapse
Affiliation(s)
- Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway
| | - Marius WiderØe
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marcus Bäck
- Department of Physics, Chemistry and Biology, Division of Chemistry, Linköping University, Linköping, Sweden
| | - K. Peter R. Nilsson
- Department of Physics, Chemistry and Biology, Division of Chemistry, Linköping University, Linköping, Sweden
| | - Per Hammarström
- Department of Physics, Chemistry and Biology, Division of Chemistry, Linköping University, Linköping, Sweden
| | - Lars N.G. Nilsson
- Department of Pharmacology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Andreas K.O. Åslund
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway
| |
Collapse
|
14
|
Kirschenbaum D, Dadgar‐Kiani E, Catto F, Voigt FF, Trevisan C, Bichsel O, Shirani H, Nilsson KPR, Frontzek KJ, Paganetti P, Helmchen F, Lee JH, Aguzzi A. Whole-brain microscopy reveals distinct temporal and spatial efficacy of anti-Aβ therapies. EMBO Mol Med 2023; 15:e16789. [PMID: 36382364 PMCID: PMC9832821 DOI: 10.15252/emmm.202216789] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/18/2022] Open
Abstract
Many efforts targeting amyloid-β (Aβ) plaques for the treatment of Alzheimer's Disease thus far have resulted in failures during clinical trials. Regional and temporal heterogeneity of efficacy and dependence on plaque maturity may have contributed to these disappointing outcomes. In this study, we mapped the regional and temporal specificity of various anti-Aβ treatments through high-resolution light-sheet imaging of electrophoretically cleared brains. We assessed the effect on amyloid plaque formation and growth in Thy1-APP/PS1 mice subjected to β-secretase inhibitors, polythiophenes, or anti-Aβ antibodies. Each treatment showed unique spatiotemporal Aβ clearance, with polythiophenes emerging as a potent anti-Aβ compound. Furthermore, aligning with a spatial-transcriptomic atlas revealed transcripts that correlate with the efficacy of each Aβ therapy. As observed in this study, there is a striking dependence of specific treatments on the location and maturity of Aβ plaques. This may also contribute to the clinical trial failures of Aβ-therapies, suggesting that combinatorial regimens may be significantly more effective in clearing amyloid deposition.
Collapse
Affiliation(s)
- Daniel Kirschenbaum
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | | | - Francesca Catto
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Fabian F Voigt
- Laboratory of Neural Circuit Dynamics, Brain Research InstituteUniversity of ZurichZurichSwitzerland
- Neuroscience Center ZurichUniversity of Zurich & ETH ZurichZurichSwitzerland
| | - Chiara Trevisan
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Oliver Bichsel
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Hamid Shirani
- Division of Chemistry, Department of Physics, Chemistry and BiologyLinköping UniversityLinköpingSweden
| | - K Peter R Nilsson
- Division of Chemistry, Department of Physics, Chemistry and BiologyLinköping UniversityLinköpingSweden
| | - Karl J Frontzek
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| | - Paolo Paganetti
- Laboratory for Biomedical NeurosciencesTorricella‐TaverneNeurocenter of Southern Switzerland, Ente Cantonale OspedalieroSwitzerland
- Faculty of Biomedical NeurosciencesUniversità della Svizzera ItalianaLuganoSwitzerland
| | - Fritjof Helmchen
- Laboratory of Neural Circuit Dynamics, Brain Research InstituteUniversity of ZurichZurichSwitzerland
| | - Jin Hyung Lee
- Department of BioengineeringStanford UniversityStanfordCAUSA
- Department of Neurology and Neurological SciencesStanford UniversityStanfordCAUSA
- Department of Electrical EngineeringStanford UniversityStanfordCAUSA
- Department of NeurosurgeryStanford UniversityStanfordCAUSA
| | - Adriano Aguzzi
- Institute of NeuropathologyUniversity Hospital ZurichUniversity of ZurichZurichSwitzerland
| |
Collapse
|
15
|
Jheng CP, Lee CI. Combination of structure-based virtual screening, molecular docking and molecular dynamics approaches for the discovery of anti-prion fibril flavonoids. Front Mol Biosci 2023; 9:1088733. [PMID: 36685276 PMCID: PMC9849400 DOI: 10.3389/fmolb.2022.1088733] [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] [Received: 11/03/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Abstract
Prion diseases are a group of rare neurodegenerative diseases caused by the structural conversion of cellular prion into Scrapie prion resulting aggregated fibrils. Therapy of prion diseases has been developed for several decades, especially drug designs based on the structure of prion monomers. Unfortunately, none of the designed anti-prion drugs function well clinically. To fight against prion fibrils, a drug design based on the precise structure of mammalian prion fibrils is highly required. Fortunately, based on the advantage of newly advanced cryo-electron microscopy (cryo-EM) in the deconvolution of large complexes, three prion fibril structures were resolved in the last 2 years. Based on the cryo-EM solved prion fibril structures, we are able to find some molecules fighting against prion fibrils. Quercetin, one flavonoid molecule in the polyphenol group, has been found to disaggregate the prion fibrils in vitro. In this study, we performed the molecular docking and molecular dynamics simulation on quercetin-like molecules possessing pharmacological properties to evaluate the anti-prion ability of tested molecules. As a result, four quercetin-like molecules interact with prion fibril and decrease the β-strand content by converting some β-strands into loop and helical structures to disintegrate the existing fibril structure. The results of this study are significant in the treatment of prion diseases, and the approaches used in this study are applicable to other amyloid diseases.
Collapse
Affiliation(s)
- Cheng-Ping Jheng
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, Taiwan
| | - Cheng-I Lee
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, Taiwan,Center for Nano Bio-Detections, National Chung Cheng University, Chia-Yi, Taiwan,Center for Innovative Research on Aging Society (CIRAS), National Chung Cheng University, Chia-Yi, Taiwan,*Correspondence: Cheng-I Lee,
| |
Collapse
|
16
|
Björk L, Bäck M, Lantz L, Ghetti B, Vidal R, Klingstedt T, Nilsson KPR. Proteophenes - Amino Acid Functionalized Thiophene-based Fluorescent Ligands for Visualization of Protein Deposits in Tissue Sections with Alzheimer's Disease Pathology. Chemistry 2022; 28:e202201557. [PMID: 35950816 PMCID: PMC9643645 DOI: 10.1002/chem.202201557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Indexed: 01/11/2023]
Abstract
Protein deposits composed of specific proteins or peptides are associated with several neurodegenerative diseases and fluorescent ligands able to detect these pathological hallmarks are vital. Here, we report the synthesis of a class of thiophene-based ligands, denoted proteophenes, with different amino acid side-chain functionalities along the conjugated backbone, which display selectivity towards specific disease-associated protein aggregates in tissue sections with Alzheimer's disease (AD) pathology. The selectivity of the ligands towards AD associated pathological hallmarks, such as aggregates of the amyloid-β (Aβ) peptide or tau filamentous inclusions, was highly dependent on the chemical nature of the amino acid functionality, as well as on the location of the functionality along the pentameric thiophene backbone. Finally, the concept of synthesizing donor-acceptor-donor proteophenes with distinct photophysical properties was shown. Our findings provide the structural and functional basis for the development of new thiophene-based ligands that can be utilized for optical assignment of different aggregated proteinaceous species in tissue sections.
Collapse
Affiliation(s)
- Linnea Björk
- Department of PhysicsChemistry and BiologyLinköping University581 83LinköpingSweden
| | - Marcus Bäck
- Department of PhysicsChemistry and BiologyLinköping University581 83LinköpingSweden
| | - Linda Lantz
- Department of PhysicsChemistry and BiologyLinköping University581 83LinköpingSweden
| | - Bernardino Ghetti
- Department of Pathology and Laboratory MedicineIndiana University School of MedicineIndianapolis46202IndianaUSA
| | - Ruben Vidal
- Department of Pathology and Laboratory MedicineIndiana University School of MedicineIndianapolis46202IndianaUSA
| | - Therése Klingstedt
- Department of PhysicsChemistry and BiologyLinköping University581 83LinköpingSweden
| | - K. Peter R. Nilsson
- Department of PhysicsChemistry and BiologyLinköping University581 83LinköpingSweden
| |
Collapse
|
17
|
A conformational switch controlling the toxicity of the prion protein. Nat Struct Mol Biol 2022; 29:831-840. [PMID: 35948768 PMCID: PMC9371974 DOI: 10.1038/s41594-022-00814-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 07/06/2022] [Indexed: 11/08/2022]
Abstract
Prion infections cause conformational changes of the cellular prion protein (PrPC) and lead to progressive neurological impairment. Here we show that toxic, prion-mimetic ligands induce an intramolecular R208-H140 hydrogen bond ('H-latch'), altering the flexibility of the α2-α3 and β2-α2 loops of PrPC. Expression of a PrP2Cys mutant mimicking the H-latch was constitutively toxic, whereas a PrPR207A mutant unable to form the H-latch conferred resistance to prion infection. High-affinity ligands that prevented H-latch induction repressed prion-related neurodegeneration in organotypic cerebellar cultures. We then selected phage-displayed ligands binding wild-type PrPC, but not PrP2Cys. These binders depopulated H-latched conformers and conferred protection against prion toxicity. Finally, brain-specific expression of an antibody rationally designed to prevent H-latch formation prolonged the life of prion-infected mice despite unhampered prion propagation, confirming that the H-latch is an important reporter of prion neurotoxicity.
Collapse
|
18
|
Ni R, Chen Z, Deán-Ben XL, Voigt FF, Kirschenbaum D, Shi G, Villois A, Zhou Q, Crimi A, Arosio P, Nitsch RM, Nilsson KPR, Aguzzi A, Helmchen F, Klohs J, Razansky D. Multiscale optical and optoacoustic imaging of amyloid-β deposits in mice. Nat Biomed Eng 2022; 6:1031-1044. [PMID: 35835994 DOI: 10.1038/s41551-022-00906-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/27/2022] [Indexed: 12/26/2022]
Abstract
Deposits of amyloid-β (Aβ) in the brains of rodents can be analysed by invasive intravital microscopy on a submillimetre scale, or via whole-brain images from modalities lacking the resolution or molecular specificity to accurately characterize Aβ pathologies. Here we show that large-field multifocal illumination fluorescence microscopy and panoramic volumetric multispectral optoacoustic tomography can be combined to longitudinally assess Aβ deposits in transgenic mouse models of Alzheimer's disease. We used fluorescent Aβ-targeted probes (the luminescent conjugated oligothiophene HS-169 and the oxazine-derivative AOI987) to transcranially detect Aβ deposits in the cortex of APP/PS1 and arcAβ mice with single-plaque resolution (8 μm) and across the whole brain (including the hippocampus and the thalamus, which are inaccessible by conventional intravital microscopy) at sub-150 μm resolutions. Two-photon microscopy, light-sheet microscopy and immunohistochemistry of brain-tissue sections confirmed the specificity and regional distributions of the deposits. High-resolution multiscale optical and optoacoustic imaging of Aβ deposits across the entire brain in rodents thus facilitates the in vivo study of Aβ accumulation by brain region and by animal age and strain.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Zhenyue Chen
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Xosé Luís Deán-Ben
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Fabian F Voigt
- Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Brain Research Institute, University of Zurich, Zurich, Switzerland
| | | | - Gloria Shi
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Alessia Villois
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Quanyu Zhou
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Alessandro Crimi
- Institute of Neuropathology, Universitätsspital Zurich, Zurich, Switzerland
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - K Peter R Nilsson
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Adriano Aguzzi
- Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Neuropathology, Universitätsspital Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland. .,Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.
| | - Daniel Razansky
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland. .,Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland. .,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Teruya K, Doh-Ura K. Therapeutic development of polymers for prion disease. Cell Tissue Res 2022; 392:349-365. [PMID: 35307792 DOI: 10.1007/s00441-022-03604-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/24/2022] [Indexed: 12/20/2022]
Abstract
Prion diseases, also known as transmissible spongiform encephalopathies, are caused by the accumulation of abnormal isoforms of the prion protein (scrapie isoform of the prion protein, PrPSc) in the central nervous system. Many compounds with anti-prion activities have been found using in silico screening, in vitro models, persistently prion-infected cell models, and prion-infected rodent models. Some of these compounds include several types of polymers. Although the inhibition or removal of PrPSc production is the main target of therapy, the unique features of prions, namely protein aggregation and assembly accompanied by steric structural transformation, may require different strategies for the development of anti-prion drugs than those for conventional therapeutics targeting enzyme inhibition, agonist ligands, or modulation of signaling. In this paper, we first overview the history of the application of polymers to prion disease research. Next, we describe the characteristics of each type of polymer with anti-prion activity. Finally, we discuss the common features of these polymers. Although drug delivery of these polymers to the brain is a challenge, they are useful not only as leads for therapeutic drugs but also as tools to explore the structure of PrPSc and are indispensable for prion disease research.
Collapse
Affiliation(s)
- Kenta Teruya
- Department of Neurochemistry, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| | - Katsumi Doh-Ura
- Department of Neurochemistry, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan.
| |
Collapse
|
20
|
Mukherjee A, Al-Lahham R, Corkins ME, Samanta S, Schmeichel AM, Singer W, Low PA, Govindaraju T, Soto C. Identification of Multicolor Fluorescent Probes for Heterogeneous Aβ Deposits in Alzheimer's Disease. Front Aging Neurosci 2022; 13:802614. [PMID: 35185519 PMCID: PMC8852231 DOI: 10.3389/fnagi.2021.802614] [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: 10/26/2021] [Accepted: 12/27/2021] [Indexed: 11/21/2022] Open
Abstract
Accumulation of amyloid-beta (Aβ) into amyloid plaques and hyperphosphorylated tau into neurofibrillary tangles (NFTs) are pathological hallmarks of Alzheimer's disease (AD). There is a significant intra- and inter-individual variability in the morphology and conformation of Aβ aggregates, which may account in part for the extensive clinical and pathophysiological heterogeneity observed in AD. In this study, we sought to identify an array of fluorescent dyes to specifically probe Aβ aggregates, in an effort to address their diversity. We screened a small library of fluorescent probes and identified three benzothiazole-coumarin derivatives that stained both vascular and parenchymal Aβ deposits in AD brain sections. The set of these three dyes allowed the visualization of Aβ deposits in three different colors (blue, green and far-red). Importantly, two of these dyes specifically stained Aβ deposits with no apparent staining of hyperphosphorylated tau or α-synuclein deposits. Furthermore, this set of dyes demonstrated differential interactions with distinct types of Aβ deposits present in the same subject. Aβ aggregate-specific dyes identified in this study have the potential to be further developed into Aβ imaging probes for the diagnosis of AD. In addition, the far-red dye we identified in this study may serve as an imaging probe for small animal imaging of Aβ pathology. Finally, these dyes in combination may help us advance our understanding of the relation between the various Aβ deposits and the clinical diversity observed in AD.
Collapse
Affiliation(s)
- Abhisek Mukherjee
- Department of Neurology, Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Rabab Al-Lahham
- Department of Neurology, Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Mark E. Corkins
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Sourav Samanta
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru, India
| | | | - Wolfgang Singer
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Phillip A. Low
- Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Bengaluru, India
| | - Claudio Soto
- Department of Neurology, Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
21
|
Roterman I, Stapor K, Gądek K, Gubała T, Nowakowski P, Fabian P, Konieczny L. On the Dependence of Prion and Amyloid Structure on the Folding Environment. Int J Mol Sci 2021; 22:ijms222413494. [PMID: 34948291 PMCID: PMC8707753 DOI: 10.3390/ijms222413494] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 01/22/2023] Open
Abstract
Currently available analyses of amyloid proteins reveal the necessity of the existence of radical structural changes in amyloid transformation processes. The analysis carried out in this paper based on the model called fuzzy oil drop (FOD) and its modified form (FOD-M) allows quantifying the role of the environment, particularly including the aquatic environment. The starting point and basis for the present presentation is the statement about the presence of two fundamentally different methods of organizing polypeptides into ordered conformations—globular proteins and amyloids. The present study shows the source of the differences between these two paths resulting from the specificity of the external force field coming from the environment, including the aquatic and hydrophobic one. The water environment expressed in the fuzzy oil drop model using the 3D Gauss function directs the folding process towards the construction of a micelle-like system with a hydrophobic core in the central part and the exposure of polarity on the surface. The hydrophobicity distribution of membrane proteins has the opposite characteristic: Exposure of hydrophobicity at the surface of the membrane protein with an often polar center (as in the case of ion channels) is expected. The structure of most proteins is influenced by a more or less modified force field generated by water through the appropriate presence of a non-polar (membrane-like) environment. The determination of the proportion of a factor different from polar water enables the assessment of the protein status by indicating factors favoring the structure it represents.
Collapse
Affiliation(s)
- Irena Roterman
- Department of Bioinformatics and Telemedicine, Jagiellonian University Medical College, 31-034 Kopernika 7, 30-688 Krakow, Poland
- Correspondence:
| | - Katarzyna Stapor
- Department of Applied Informatics, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland;
| | - Krzysztof Gądek
- Sano Centre for Computation Medicine, Czarnowiejska 36, 30-054 Kraków, Poland; (K.G.); (T.G.); (P.N.)
| | - Tomasz Gubała
- Sano Centre for Computation Medicine, Czarnowiejska 36, 30-054 Kraków, Poland; (K.G.); (T.G.); (P.N.)
| | - Piotr Nowakowski
- Sano Centre for Computation Medicine, Czarnowiejska 36, 30-054 Kraków, Poland; (K.G.); (T.G.); (P.N.)
| | - Piotr Fabian
- Department of Algorithmics and Software, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland;
| | - Leszek Konieczny
- Department of Medical Biochemistry, Jagiellonian University Medical College, 31-034 Kopernika 7, 31-034 Krakow, Poland;
| |
Collapse
|
22
|
Liu H, Kim C, Haldiman T, Sigurdson CJ, Nyström S, Nilsson KPR, Cohen ML, Wisniewski T, Hammarström P, Safar JG. Distinct conformers of amyloid beta accumulate in the neocortex of patients with rapidly progressive Alzheimer's disease. J Biol Chem 2021; 297:101267. [PMID: 34599965 PMCID: PMC8531671 DOI: 10.1016/j.jbc.2021.101267] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/03/2022] Open
Abstract
Amyloid beta (Aβ) deposition in the neocortex is a major hallmark of Alzheimer's disease (AD), but the extent of deposition does not readily explain phenotypic diversity and rate of disease progression. The prion strain-like model of disease heterogeneity suggests the existence of different conformers of Aβ. We explored this paradigm using conformation-dependent immunoassay (CDI) for Aβ and conformation-sensitive luminescent conjugated oligothiophenes (LCOs) in AD cases with variable progression rates. Mapping the Aβ conformations in the frontal, occipital, and temporal regions in 20 AD patients with CDI revealed extensive interindividual and anatomical diversity in the structural organization of Aβ with the most significant differences in the temporal cortex of rapidly progressive AD. The fluorescence emission spectra collected in situ from Aβ plaques in the same regions demonstrated considerable diversity of spectral characteristics of two LCOs-quatroformylthiophene acetic acid and heptaformylthiophene acetic acid. Heptaformylthiophene acetic acid detected a wider range of Aβ deposits, and both LCOs revealed distinct spectral attributes of diffuse and cored plaques in the temporal cortex of rapidly and slowly progressive AD and less frequent and discernible differences in the frontal and occipital cortex. These and CDI findings indicate a major conformational diversity of Aβ accumulating in the neocortex, with the most notable differences in temporal cortex of cases with shorter disease duration, and implicate distinct Aβ conformers (strains) in the rapid progression of AD.
Collapse
Affiliation(s)
- He Liu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Chae Kim
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Tracy Haldiman
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Christina J Sigurdson
- Department of Pathology, University of California, San Diego, La Jolla, California, USA; Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Sofie Nyström
- Department of Physics, Chemistry, and Biology, Linköping University, Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry, and Biology, Linköping University, Linköping, Sweden
| | - Mark L Cohen
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA; National Prion Disease Pathology Surveillance Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Thomas Wisniewski
- Centre for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, New York, USA; Department of Psychiatry, New York University School of Medicine, New York, New York, USA
| | - Per Hammarström
- Department of Physics, Chemistry, and Biology, Linköping University, Linköping, Sweden
| | - Jiri G Safar
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA; Department of Neurology, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
23
|
Roterman I, Stapor K, Fabian P, Konieczny L. In Silico Modeling of the Influence of Environment on Amyloid Folding Using FOD-M Model. Int J Mol Sci 2021; 22:10587. [PMID: 34638925 PMCID: PMC8508659 DOI: 10.3390/ijms221910587] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 11/17/2022] Open
Abstract
The role of the environment in amyloid formation based on the fuzzy oil drop model (FOD) is discussed here. This model assumes that the hydrophobicity distribution within a globular protein is consistent with a 3D Gaussian (3DG) distribution. Such a distribution is interpreted as the idealized effect of the presence of a polar solvent-water. A chain with a sequence of amino acids (which are bipolar molecules) determined by evolution recreates a micelle-like structure with varying accuracy. The membrane, which is a specific environment with opposite characteristics to the polar aquatic environment, directs the hydrophobic residues towards the surface. The modification of the FOD model to the FOD-M form takes into account the specificity of the cell membrane. It consists in "inverting" the 3DG distribution (complementing the Gaussian distribution), which expresses the exposure of hydrophobic residues on the surface. It turns out that the influence of the environment for any protein (soluble or membrane-anchored) is the result of a consensus factor expressing the participation of the polar environment and the "inverted" environment. The ratio between the proportion of the aqueous and the "reversed" environment turns out to be a characteristic property of a given protein, including amyloid protein in particular. The structure of amyloid proteins has been characterized in the context of prion, intrinsically disordered, and other non-complexing proteins to cover a wider spectrum of molecules with the given characteristics based on the FOD-M model.
Collapse
Affiliation(s)
- Irena Roterman
- Department of Bioinformatics and Telemedicine, Medical College, Jagiellonian University, Medyczna 7, 30-688 Kraków, Poland
| | - Katarzyna Stapor
- Institute of Computer Science, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland; (K.S.); (P.F.)
| | - Piotr Fabian
- Institute of Computer Science, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland; (K.S.); (P.F.)
| | - Leszek Konieczny
- Chair of Medical Biochemistry, Medical College, Jagiellonian University, Kopernika 7, 31-034 Kraków, Poland;
| |
Collapse
|
24
|
Ozono H, Ishikawa T. Visualization of the Interfacial Electrostatic Complementarity: A Method for Analysis of Protein-Protein Interaction Based on Ab Initio Quantum Chemical Calculations. J Chem Theory Comput 2021; 17:5600-5610. [PMID: 34432447 DOI: 10.1021/acs.jctc.1c00475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In this study, we report a method for analyzing the protein-protein interaction based on ab initio quantum chemical calculations, which we refer to as "Visualization of the interfacial electrostatic complementarity (VIINEC)." This method visually provides the electrostatic complementarity at the protein-protein interface; in addition, the ratio of the attractive interaction is calculated. Illustrative calculations revealed that VIINEC could successfully quantify the electronic induced fit owing complex formation, which was responsible for 5%-10% of the total electrostatic complementarity. Furthermore, the contribution of each amino acid to the electrostatic complementarity was evaluated, providing useful information for various applications, including rational antibody designs. Interestingly, a part of the mechanism causing the specificity of the protein-protein bindings was also demonstrated using VIINEC. This is an important achievement of this study because the specificity of the biomolecular interactions is essential for biological functions.
Collapse
Affiliation(s)
- Hiroki Ozono
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima, Kagoshima 890-0065, Japan
| | - Takeshi Ishikawa
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima, Kagoshima 890-0065, Japan
| |
Collapse
|
25
|
Abstract
Prion diseases are neurodegenerative disorders caused by conformational conversion of the cellular prion protein (PrPC) into scrapie prion protein (PrPSc). As the main component of prion, PrPSc acts as an infectious template that recruits and converts normal cellular PrPC into its pathogenic, misfolded isoform. Intriguingly, the phenomenon of prionoid, or prion-like, spread has also been observed in many other disease-associated proteins, such as amyloid β (Aβ), tau and α-synuclein. This Cell Science at a Glance and the accompanying poster highlight recently described physiological roles of prion protein and the advanced understanding of pathogenesis of prion disease they have afforded. Importantly, prion protein may also be involved in the pathogenesis of other neurodegenerative disorders such as Alzheimer's and Parkinson's disease. Therapeutic studies of prion disease have also exploited novel strategies to combat these devastating diseases. Future studies on prion protein and prion disease will deepen our understanding of the pathogenesis of a broad spectrum of neurodegenerative conditions.
Collapse
Affiliation(s)
- Caihong Zhu
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zürich, Zürich, CH-8091, Switzerland
| |
Collapse
|
26
|
Klingstedt T, Shirani H, Ghetti B, Vidal R, R. Nilsson KP. Thiophene-Based Optical Ligands That Selectively Detect Aβ Pathology in Alzheimer's Disease. Chembiochem 2021; 22:2568-2581. [PMID: 34101954 PMCID: PMC8409278 DOI: 10.1002/cbic.202100199] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/04/2021] [Indexed: 11/25/2022]
Abstract
In several neurodegenerative diseases, the presence of aggregates of specific proteins in the brain is a significant pathological hallmark; thus, developing ligands able to bind to the aggregated proteins is essential for any effort related to imaging and therapeutics. Here we report the synthesis of thiophene-based ligands containing nitrogen heterocycles. The ligands selectively recognized amyloid-β (Aβ) aggregates in brain tissue from individuals diagnosed neuropathologically as having Alzheimer's disease (AD). The selectivity for Aβ was dependent on the position of nitrogen in the heterocyclic compounds, and the ability to bind Aβ was shown to be reduced when introducing anionic substituents on the thiophene backbone. Our findings provide the structural and functional basis for the development of ligands that can differentiate between aggregated proteinaceous species comprised of distinct proteins. These ligands might also be powerful tools for studying the pathogenesis of Aβ aggregation and for designing molecules for imaging of Aβ pathology.
Collapse
Affiliation(s)
- Therése Klingstedt
- Department of PhysicsChemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| | - Hamid Shirani
- Department of PhysicsChemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| | - Bernardino Ghetti
- Department of Pathology and Laboratory MedicineIndiana University School of MedicineIndianapolis46202IndianaUSA
| | - Ruben Vidal
- Department of Pathology and Laboratory MedicineIndiana University School of MedicineIndianapolis46202IndianaUSA
| | - K. Peter R. Nilsson
- Department of PhysicsChemistry and BiologyLinköping UniversitySE-581 83LinköpingSweden
| |
Collapse
|
27
|
Carta M, Aguzzi A. Molecular foundations of prion strain diversity. Curr Opin Neurobiol 2021; 72:22-31. [PMID: 34416480 DOI: 10.1016/j.conb.2021.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Despite being caused by a single protein, prion diseases are strikingly heterogenous. Individual prion variants, known as strains, possess distinct biochemical properties, form aggregates with characteristic morphologies and preferentially seed certain brain regions, causing markedly different disease phenotypes. Strain diversity is determined by protein structure, post-translational modifications and the presence of extracellular matrix components, with single amino acid substitutions or altered protein glycosylation exerting dramatic effects. Here, we review recent advances in the study of prion strains and discuss how a deeper knowledge of the molecular origins of strain heterogeneity is providing a foundation for the development of anti-prion therapeutics.
Collapse
Affiliation(s)
- Manfredi Carta
- Institute of Neuropathology, University Hospital of Zurich, University of Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital of Zurich, University of Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland.
| |
Collapse
|
28
|
Lakkaraju AKK, Frontzek K, Lemes E, Herrmann U, Losa M, Marpakwar R, Aguzzi A. Loss of PIKfyve drives the spongiform degeneration in prion diseases. EMBO Mol Med 2021; 13:e14714. [PMID: 34291577 PMCID: PMC8518562 DOI: 10.15252/emmm.202114714] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 11/21/2022] Open
Abstract
Brain‐matter vacuolation is a defining trait of all prion diseases, yet its cause is unknown. Here, we report that prion infection and prion‐mimetic antibodies deplete the phosphoinositide kinase PIKfyve—which controls endolysosomal maturation—from mouse brains, cultured cells, organotypic brain slices, and brains of Creutzfeldt‐Jakob disease victims. We found that PIKfyve is acylated by the acyltransferases zDHHC9 and zDHHC21, whose juxtavesicular topology is disturbed by prion infection, resulting in PIKfyve deacylation and rapid degradation, as well as endolysosomal hypertrophy and activation of TFEB‐dependent lysosomal enzymes. A protracted unfolded protein response (UPR), typical of prion diseases, also induced PIKfyve deacylation and degradation. Conversely, UPR antagonists restored PIKfyve levels in prion‐infected cells. Overexpression of zDHHC9 and zDHHC21, administration of the antiprion polythiophene LIN5044, or supplementation with the PIKfyve reaction product PI(3,5)P2 suppressed prion‐induced vacuolation and restored lysosomal homeostasis. Thus, PIKfyve emerges as a central mediator of vacuolation and neurotoxicity in prion diseases.
Collapse
Affiliation(s)
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Emina Lemes
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Uli Herrmann
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - Marco Losa
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | | | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| |
Collapse
|
29
|
Lutter L, Aubrey LD, Xue WF. On the Structural Diversity and Individuality of Polymorphic Amyloid Protein Assemblies. J Mol Biol 2021; 433:167124. [PMID: 34224749 DOI: 10.1016/j.jmb.2021.167124] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/20/2021] [Accepted: 06/26/2021] [Indexed: 12/24/2022]
Abstract
The prediction of highly ordered three-dimensional structures of amyloid protein fibrils from the amino acid sequences of their monomeric self-assembly precursors constitutes a challenging and unresolved aspect of the classical protein folding problem. Because of the polymorphic nature of amyloid assembly whereby polypeptide chains of identical amino acid sequences under identical conditions are capable of self-assembly into a spectrum of different fibril structures, the prediction of amyloid structures from an amino acid sequence requires a detailed and holistic understanding of its assembly free energy landscape. The full extent of the structure space accessible to the cross-β molecular architecture of amyloid must also be resolved. Here, we review the current understanding of the diversity and the individuality of amyloid structures, and how the polymorphic landscape of amyloid links to biology and disease phenotypes. We present a comprehensive review of structural models of amyloid fibrils derived by cryo-EM, ssNMR and AFM to date, and discuss the challenges ahead for resolving the structural basis and the biological consequences of polymorphic amyloid assemblies.
Collapse
Affiliation(s)
- Liisa Lutter
- School of Biosciences, Division of Natural Sciences, University of Kent, CT2 7NJ Canterbury, UK
| | - Liam D Aubrey
- School of Biosciences, Division of Natural Sciences, University of Kent, CT2 7NJ Canterbury, UK
| | - Wei-Feng Xue
- School of Biosciences, Division of Natural Sciences, University of Kent, CT2 7NJ Canterbury, UK.
| |
Collapse
|
30
|
Lewkowicz E, Jayaraman S, Gursky O. Protein Amyloid Cofactors: Charged Side-Chain Arrays Meet Their Match? Trends Biochem Sci 2021; 46:626-629. [PMID: 34210544 DOI: 10.1016/j.tibs.2021.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/05/2021] [Accepted: 05/08/2021] [Indexed: 12/25/2022]
Abstract
Recent advances in high-resolution structural studies of protein amyloids have revealed parallel in-register cross-β-sheets with periodic arrays of closely spaced identical residues. What do these structures tell us about the mechanisms of action of common amyloid-promoting factors, such as heparan sulfate (HS), nucleic acids, polyphosphates, anionic phospholipids, and acidic pH?
Collapse
Affiliation(s)
- Emily Lewkowicz
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Shobini Jayaraman
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA
| | - Olga Gursky
- Department of Physiology and Biophysics, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
31
|
Shi Y, Murzin AG, Falcon B, Epstein A, Machin J, Tempest P, Newell KL, Vidal R, Garringer HJ, Sahara N, Higuchi M, Ghetti B, Jang MK, Scheres SHW, Goedert M. Cryo-EM structures of tau filaments from Alzheimer's disease with PET ligand APN-1607. Acta Neuropathol 2021; 141:697-708. [PMID: 33723967 PMCID: PMC8043864 DOI: 10.1007/s00401-021-02294-3] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
Tau and Aβ assemblies of Alzheimer’s disease (AD) can be visualized in living subjects using positron emission tomography (PET). Tau assemblies comprise paired helical and straight filaments (PHFs and SFs). APN-1607 (PM-PBB3) is a recently described PET ligand for AD and other tau proteinopathies. Since it is not known where in the tau folds PET ligands bind, we used electron cryo-microscopy (cryo-EM) to determine the binding sites of APN-1607 in the Alzheimer fold. We identified two major sites in the β-helix of PHFs and SFs and a third major site in the C-shaped cavity of SFs. In addition, we report that tau filaments from posterior cortical atrophy (PCA) and primary age-related tauopathy (PART) are identical to those from AD. In support, fluorescence labelling showed binding of APN-1607 to intraneuronal inclusions in AD, PART and PCA. Knowledge of the binding modes of APN-1607 to tau filaments may lead to the development of new ligands with increased specificity and binding activity. We show that cryo-EM can be used to identify the binding sites of small molecules in amyloid filaments.
Collapse
|
32
|
Milardi D, Gazit E, Radford SE, Xu Y, Gallardo RU, Caflisch A, Westermark GT, Westermark P, Rosa CL, Ramamoorthy A. Proteostasis of Islet Amyloid Polypeptide: A Molecular Perspective of Risk Factors and Protective Strategies for Type II Diabetes. Chem Rev 2021; 121:1845-1893. [PMID: 33427465 PMCID: PMC10317076 DOI: 10.1021/acs.chemrev.0c00981] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The possible link between hIAPP accumulation and β-cell death in diabetic patients has inspired numerous studies focusing on amyloid structures and aggregation pathways of this hormone. Recent studies have reported on the importance of early oligomeric intermediates, the many roles of their interactions with lipid membrane, pH, insulin, and zinc on the mechanism of aggregation of hIAPP. The challenges posed by the transient nature of amyloid oligomers, their structural heterogeneity, and the complex nature of their interaction with lipid membranes have resulted in the development of a wide range of biophysical and chemical approaches to characterize the aggregation process. While the cellular processes and factors activating hIAPP-mediated cytotoxicity are still not clear, it has recently been suggested that its impaired turnover and cellular processing by proteasome and autophagy may contribute significantly toward toxic hIAPP accumulation and, eventually, β-cell death. Therefore, studies focusing on the restoration of hIAPP proteostasis may represent a promising arena for the design of effective therapies. In this review we discuss the current knowledge of the structures and pathology associated with hIAPP self-assembly and point out the opportunities for therapy that a detailed biochemical, biophysical, and cellular understanding of its aggregation may unveil.
Collapse
Affiliation(s)
- Danilo Milardi
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Via P. Gaifami 18, 95126 Catania, Italy
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Yong Xu
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Rodrigo U Gallardo
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zürich, Zürich CH-8057, Switzerland
| | - Gunilla T Westermark
- Department of Medical Cell Biology, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Per Westermark
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Carmelo La Rosa
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Ayyalusamy Ramamoorthy
- Biophysics, Department of Chemistry, Biomedical Engineering, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan 41809-1055, United States
| |
Collapse
|
33
|
Cingolani M, Mummolo L, Lugli F, Zaffagnini M, Genovese D. Protein aggregation detection with fluorescent macromolecular and nanostructured probes: challenges and opportunities. NEW J CHEM 2021. [DOI: 10.1039/d1nj01606g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanoprobes based on various nanomaterials, polymers or AIEgens are overcoming previous limitations for diagnosis and therapy of early-stage protein aggregation.
Collapse
Affiliation(s)
- Matteo Cingolani
- Dipartimento di Chimica “Giacomo Ciamician”
- Università di Bologna
- 40126 Bologna
- Italy
| | - Liviana Mummolo
- Dipartimento di Chimica “Giacomo Ciamician”
- Università di Bologna
- 40126 Bologna
- Italy
| | - Francesca Lugli
- Dipartimento di Chimica “Giacomo Ciamician”
- Università di Bologna
- 40126 Bologna
- Italy
| | - Mirko Zaffagnini
- Dipartimento di Farmacia e Biotecnologia
- Università di Bologna
- 40126 Bologna
- Italy
| | - Damiano Genovese
- Dipartimento di Chimica “Giacomo Ciamician”
- Università di Bologna
- 40126 Bologna
- Italy
| |
Collapse
|
34
|
Mustazza C, Sbriccoli M, Minosi P, Raggi C. Small Molecules with Anti-Prion Activity. Curr Med Chem 2020; 27:5446-5479. [PMID: 31560283 DOI: 10.2174/0929867326666190927121744] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 08/08/2019] [Accepted: 09/05/2019] [Indexed: 01/20/2023]
Abstract
Prion pathologies are fatal neurodegenerative diseases caused by the misfolding of the physiological Prion Protein (PrPC) into a β-structure-rich isoform called PrPSc. To date, there is no available cure for prion diseases and just a few clinical trials have been carried out. The initial approach in the search of anti-prion agents had PrPSc as a target, but the existence of different prion strains arising from alternative conformations of PrPSc, limited the efficacy of the ligands to a straindependent ability. That has shifted research to PrPC ligands, which either act as chaperones, by stabilizing the native conformation, or inhibit its interaction with PrPSc. The role of transition-metal mediated oxidation processes in prion misfolding has also been investigated. Another promising approach is the indirect action via other cellular targets, like membrane domains or the Protein- Folding Activity of Ribosomes (PFAR). Also, new prion-specific high throughput screening techniques have been developed. However, so far no substance has been found to be able to extend satisfactorily survival time in animal models of prion diseases. This review describes the main features of the Structure-Activity Relationship (SAR) of the various chemical classes of anti-prion agents.
Collapse
Affiliation(s)
- Carlo Mustazza
- National Centre for Control and Evaluation of Medicines, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marco Sbriccoli
- Department of Neurosciences, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Paola Minosi
- National Centre for Drug Research and Evaluation, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Carla Raggi
- National Centre for Control and Evaluation of Medicines, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
35
|
Minikel EV, Zhao HT, Le J, O'Moore J, Pitstick R, Graffam S, Carlson GA, Kavanaugh MP, Kriz J, Kim JB, Ma J, Wille H, Aiken J, McKenzie D, Doh-Ura K, Beck M, O'Keefe R, Stathopoulos J, Caron T, Schreiber SL, Carroll JB, Kordasiewicz HB, Cabin DE, Vallabh SM. Prion protein lowering is a disease-modifying therapy across prion disease stages, strains and endpoints. Nucleic Acids Res 2020; 48:10615-10631. [PMID: 32776089 PMCID: PMC7641729 DOI: 10.1093/nar/gkaa616] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/23/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Lowering of prion protein (PrP) expression in the brain is a genetically validated therapeutic hypothesis in prion disease. We recently showed that antisense oligonucleotide (ASO)-mediated PrP suppression extends survival and delays disease onset in intracerebrally prion-infected mice in both prophylactic and delayed dosing paradigms. Here, we examine the efficacy of this therapeutic approach across diverse paradigms, varying the dose and dosing regimen, prion strain, treatment timepoint, and examining symptomatic, survival, and biomarker readouts. We recapitulate our previous findings with additional PrP-targeting ASOs, and demonstrate therapeutic benefit against four additional prion strains. We demonstrate that <25% PrP suppression is sufficient to extend survival and delay symptoms in a prophylactic paradigm. Rise in both neuroinflammation and neuronal injury markers can be reversed by a single dose of PrP-lowering ASO administered after the detection of pathological change. Chronic ASO-mediated suppression of PrP beginning at any time up to early signs of neuropathology confers benefit similar to constitutive heterozygous PrP knockout. Remarkably, even after emergence of frank symptoms including weight loss, a single treatment prolongs survival by months in a subset of animals. These results support ASO-mediated PrP lowering, and PrP-lowering therapeutics in general, as a promising path forward against prion disease.
Collapse
Affiliation(s)
- Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Prion Alliance, Cambridge, MA, 02139, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Hien T Zhao
- Ionis Pharmaceuticals Inc, Carlsbad, CA 92010, USA
| | - Jason Le
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jill O'Moore
- McLaughlin Research Institute, Great Falls, MT 59405, USA
| | - Rose Pitstick
- McLaughlin Research Institute, Great Falls, MT 59405, USA
| | | | | | | | - Jasna Kriz
- Cervo Brain Research Center, Université Laval, Québec, QC G1J 2G3, Canada
| | | | - Jiyan Ma
- Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Holger Wille
- University of Alberta, Edmonton, AB T6G 2M8, Canada
| | - Judd Aiken
- University of Alberta, Edmonton, AB T6G 2M8, Canada
| | | | - Katsumi Doh-Ura
- Department of Neurochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Matthew Beck
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rhonda O'Keefe
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Tyler Caron
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Stuart L Schreiber
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | - Sonia M Vallabh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Prion Alliance, Cambridge, MA, 02139, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
36
|
Sevillano AM, Aguilar-Calvo P, Kurt TD, Lawrence JA, Soldau K, Nam TH, Schumann T, Pizzo DP, Nyström S, Choudhury B, Altmeppen H, Esko JD, Glatzel M, Nilsson KPR, Sigurdson CJ. Prion protein glycans reduce intracerebral fibril formation and spongiosis in prion disease. J Clin Invest 2020; 130:1350-1362. [PMID: 31985492 DOI: 10.1172/jci131564] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022] Open
Abstract
Posttranslational modifications (PTMs) are common among proteins that aggregate in neurodegenerative disease, yet how PTMs impact the aggregate conformation and disease progression remains unclear. By engineering knockin mice expressing prion protein (PrP) lacking 2 N-linked glycans (Prnp180Q/196Q), we provide evidence that glycans reduce spongiform degeneration and hinder plaque formation in prion disease. Prnp180Q/196Q mice challenged with 2 subfibrillar, non-plaque-forming prion strains instead developed plaques highly enriched in ADAM10-cleaved PrP and heparan sulfate (HS). Intriguingly, a third strain composed of intact, glycophosphatidylinositol-anchored (GPI-anchored) PrP was relatively unchanged, forming diffuse, HS-deficient deposits in both the Prnp180Q/196Q and WT mice, underscoring the pivotal role of the GPI-anchor in driving the aggregate conformation and disease phenotype. Finally, knockin mice expressing triglycosylated PrP (Prnp187N) challenged with a plaque-forming prion strain showed a phenotype reversal, with a striking disease acceleration and switch from plaques to predominantly diffuse, subfibrillar deposits. Our findings suggest that the dominance of subfibrillar aggregates in prion disease is due to the replication of GPI-anchored prions, with fibrillar plaques forming from poorly glycosylated, GPI-anchorless prions that interact with extracellular HS. These studies provide insight into how PTMs impact PrP interactions with polyanionic cofactors, and highlight PTMs as a major force driving the prion disease phenotype.
Collapse
Affiliation(s)
| | | | - Timothy D Kurt
- Department of Pathology, UCSD, La Jolla, California, USA
| | | | - Katrin Soldau
- Department of Pathology, UCSD, La Jolla, California, USA
| | - Thu H Nam
- Department of Pathology, UCSD, La Jolla, California, USA
| | | | - Donald P Pizzo
- Department of Pathology, UCSD, La Jolla, California, USA
| | - Sofie Nyström
- Department of Physics, Chemistry, and Biology, Linköping University, Linköping, Sweden
| | - Biswa Choudhury
- Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA
| | - Hermann Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jeffrey D Esko
- Department of Cellular and Molecular Medicine, UCSD, La Jolla, California, USA
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - K Peter R Nilsson
- Department of Physics, Chemistry, and Biology, Linköping University, Linköping, Sweden
| | - Christina J Sigurdson
- Department of Pathology, UCSD, La Jolla, California, USA.,Department of Medicine, UCSD, La Jolla, California, USA.,Department of Pathology, Immunology, and Microbiology, UCD, Davis, California, USA
| |
Collapse
|
37
|
Abstract
Prions were initially discovered in studies of scrapie, a transmissible neurodegenerative disease (ND) of sheep and goats thought to be caused by slow viruses. Once scrapie was transmitted to rodents, it was discovered that the scrapie pathogen resisted inactivation by procedures that modify nucleic acids. Eventually, this novel pathogen proved to be a protein of 209 amino acids, which is encoded by a chromosomal gene. After the absence of a nucleic acid within the scrapie agent was established, the mechanism of infectivity posed a conundrum and eliminated a hypothetical virus. Subsequently, the infectious scrapie prion protein (PrPSc) enriched for β-sheet was found to be generated from the cellular prion protein (PrPC) that is predominantly α-helical. The post-translational process that features in nascent prion formation involves a templated conformational change in PrPC that results in an infectious copy of PrPSc. Thus, prions are proteins that adopt alternative conformations, which are self-propagating and found in organisms ranging from yeast to humans. Prions have been found in both Alzheimer's (AD) and Parkinson's (PD) diseases. Mutations in APP and α-synuclein genes have been shown to cause familial AD and PD. Recently, AD was found to be a double prion disorder: both Aβ and tau prions feature in this ND. Increasing evidence argues for α-synuclein prions as the cause of PD, multiple system atrophy, and Lewy body dementia.
Collapse
|
38
|
Recent developments in antibody therapeutics against prion disease. Emerg Top Life Sci 2020; 4:169-173. [PMID: 32633322 DOI: 10.1042/etls20200002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/15/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022]
Abstract
Preclinical evidence indicates that prion diseases can respond favorably to passive immunotherapy. However, certain antibodies to the cellular prion protein PrPC can be toxic. Comprehensive studies of structure-function relationships have revealed that the flexible amino-terminal tail of PrPC is instrumental for mediating prion toxicity. In a first-in-human study, an anti-prion antibody has been recently administered to patients diagnosed with sporadic Creutzfeldt-Jakob's disease, the most prevalent human prion disease. Moreover, large-scale serosurveys have mapped the prevalence of naturally occurring human anti-prion autoantibodies in health and disease. Here, we provide a perspective on the limitations and opportunities of therapeutic anti-prion antibodies.
Collapse
|
39
|
Stepanchuk A, Tahir W, Nilsson KPR, Schatzl HM, Stys PK. Early detection of prion protein aggregation with a fluorescent pentameric oligothiophene probe using spectral confocal microscopy. J Neurochem 2020; 156:1033-1048. [PMID: 32799317 DOI: 10.1111/jnc.15148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/14/2020] [Accepted: 07/31/2020] [Indexed: 11/30/2022]
Abstract
Misfolding of the prion protein (PrP) and templating of its pathological conformation onto cognate proteins causes a number of lethal disorders of central nervous system in humans and animals, such as Creutzfeldt-Jacob disease, chronic wasting disease and bovine spongiform encephalopathy. Structural rearrangement of PrPC into PrPSc promotes aggregation of misfolded proteins into β-sheet-rich fibrils, which can be visualized by conformationally sensitive fluorescent probes. Early detection of prion misfolding and deposition might provide useful insights into its pathophysiology. Pentameric formyl thiophene acetic acid (pFTAA) is a novel amyloid probe that was shown to sensitively detect various misfolded proteins, including PrP. Here, we compared sensitivity of pFTAA staining and spectral microscopy with conventional methods of prion detection in mouse brains infected with mouse-adapted 22L prions. pFTAA bound to prion deposits in mouse brain sections exhibited a red-shifted fluorescence emission spectrum, which quantitatively increased with disease progression. Small prion deposits were detected as early as 50 days post-inoculation, well before appearance of clinical signs. Moreover, we detected significant spectral shifts in the greater brain parenchyma as early as 25 days post-inoculation, rivaling the most sensitive conventional method (real-time quaking-induced conversion). These results showcase the potential of pFTAA staining combined with spectral imaging for screening of prion-infected tissue. Not only does this method have comparable sensitivity to established techniques, it is faster and technically simpler. Finally, this readout provides valuable information about the spatial distribution of prion aggregates across tissue in the earliest stages of infection, potentially providing valuable pathophysiological insight into prion transmission.
Collapse
Affiliation(s)
- Anastasiia Stepanchuk
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Waqas Tahir
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Hermann M Schatzl
- Calgary Prion Research Unit, Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Peter K Stys
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
40
|
Cazzaniga FA, De Luca CMG, Bistaffa E, Consonni A, Legname G, Giaccone G, Moda F. Cell-free amplification of prions: Where do we stand? PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:325-358. [PMID: 32958239 DOI: 10.1016/bs.pmbts.2020.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), atypical parkinsonisms, frontotemporal dementia (FTLD) and prion diseases are characterized by the accumulation of misfolded proteins in the central nervous system (CNS). Although the cause for the initiation of protein aggregation is not well understood, these aggregates are disease-specific. For instance, AD is characterized by the intraneuronal accumulation of tau and extracellular deposition of amyloid-β (Aβ), PD is marked by the intraneuronal accumulation of α-synuclein, many FTLD are associated with the accumulation of TDP-43 while prion diseases show aggregates of misfolded prion protein. Hence, misfolded proteins are considered disease-specific biomarkers and their identification and localization in the CNS, collected postmortem, is required for a definitive diagnosis. With the development of two innovative cell-free amplification techniques named Protein Misfolding Cyclic Amplification (PMCA) and Real-Time Quaking-Induced Conversion (RT-QuIC), traces of disease-specific biomarkers were found in CSF and other peripheral tissues (e.g., urine, blood, and olfactory mucosa) of patients with different NDs. These techniques exploit an important feature shared by many misfolded proteins, that is their ability to interact with their normally folded counterparts and force them to undergo similar structural rearrangements. Essentially, RT-QuIC and PMCA mimic in vitro the same pathological processes of protein misfolding which occur in vivo in a very rapid manner. For this reason, they have been employed for studying different aspects of protein misfolding but, overall, they seem to be very promising for the premortem diagnosis of NDs.
Collapse
Affiliation(s)
- Federico Angelo Cazzaniga
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Neurology 5 and Neuropathology, Milan, Italy
| | | | - Edoardo Bistaffa
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Neurology 5 and Neuropathology, Milan, Italy
| | - Alessandra Consonni
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Milan, Italy
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore Di Studi Avanzati (SISSA), Trieste, Italy
| | - Giorgio Giaccone
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Neurology 5 and Neuropathology, Milan, Italy
| | - Fabio Moda
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Unit of Neurology 5 and Neuropathology, Milan, Italy.
| |
Collapse
|
41
|
Colini Baldeschi A, Vanni S, Zattoni M, Legname G. Novel regulators of PrP C expression as potential therapeutic targets in prion diseases. Expert Opin Ther Targets 2020; 24:759-776. [PMID: 32631090 DOI: 10.1080/14728222.2020.1782384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Prion diseases are rare and fatal neurodegenerative disorders. The key molecular event in these disorders is the misfolding of the physiological form of the cellular prion protein, PrPC, leading to the accumulation of a pathological isoform, PrPSc, with unique features. Both isoforms share the same primary sequence, lacking detectable differences in posttranslational modification, a major hurdle for their biochemical or biophysical independent characterization. The mechanism underlying the conversion of PrPC to PrPSc is not completely understood, so finding an effective therapy to cure prion disorders is extremely challenging. AREAS COVERED This review discusses the strategies for decreasing prion replication and throws a spotlight on the relevance of PrPC in the prion accumulation process. EXPERT OPINION PrPC is the key substrate for prion pathology; hence, the most promising therapeutic approach appears to be the targeting of PrPC to block the production of the infectious isoform. The use of RNA interference and antisense oligonucleotide technologies may offer opportunities for treatment because of their success in clinical trials for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Arianna Colini Baldeschi
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore Di Studi Avanzati (SISSA) , Trieste, Italy
| | - Silvia Vanni
- Osteoncology and Rare Tumors Center, Istituto Scientifico Romagnolo per Lo Studio E La Cura Dei Tumori (IRST) IRCCS , Meldola, Italy
| | - Marco Zattoni
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore Di Studi Avanzati (SISSA) , Trieste, Italy
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore Di Studi Avanzati (SISSA) , Trieste, Italy
| |
Collapse
|
42
|
Parlak O, Richter-Dahlfors A. Bacterial Sensing and Biofilm Monitoring for Infection Diagnostics. Macromol Biosci 2020; 20:e2000129. [PMID: 32588553 DOI: 10.1002/mabi.202000129] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/01/2020] [Indexed: 12/21/2022]
Abstract
Recent insights into the rapidly emerging field of bacterial sensing and biofilm monitoring for infection diagnostics are discussed as well as recent key developments and emerging technologies in the field. Electrochemical sensing of bacteria and bacterial biofilm via synthetic, natural, and engineered recognition, as well as direct redox-sensing approaches via algorithm-based optical sensing, and tailor-made optotracing technology are discussed. These technologies are highlighted to answer the very critical question: "how can fast and accurate bacterial sensing and biofilm monitoring be achieved? Following on from that: "how can these different sensing concepts be translated for use in infection diagnostics? A central obstacle to this transformation is the absence of direct and fast analysis methods that provide high-throughput results and bio-interfaces that can control and regulate the means of communication between biological and electronic systems. Here, the overall progress made to date in building such translational efforts at the level of an individual bacterial cell to a bacterial community is discussed.
Collapse
Affiliation(s)
- Onur Parlak
- AIMES-Center for the Advancement of Integrated Medical and Engineering Science, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, SE-171 77, Sweden.,Department of Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden
| | - Agneta Richter-Dahlfors
- AIMES-Center for the Advancement of Integrated Medical and Engineering Science, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, SE-171 77, Sweden.,Department of Neuroscience, Karolinska Institutet, Stockholm, SE-171 77, Sweden.,Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, SE-100 44, Sweden
| |
Collapse
|
43
|
Zaccagnini L, Rossetti G, Tran TH, Salzano G, Gandini A, Colini Baldeschi A, Bolognesi ML, Carloni P, Legname G. In silico/in vitro screening and hit evaluation identified new phenothiazine anti-prion derivatives. Eur J Med Chem 2020; 196:112295. [DOI: 10.1016/j.ejmech.2020.112295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022]
|
44
|
Lantz L, Shirani H, Klingstedt T, Nilsson KPR. Synthesis and Characterization of Thiophene-based Donor-Acceptor-Donor Heptameric Ligands for Spectral Assignment of Polymorphic Amyloid-β Deposits. Chemistry 2020; 26:7425-7432. [PMID: 32022335 PMCID: PMC7318160 DOI: 10.1002/chem.201905612] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Indexed: 11/29/2022]
Abstract
Protein deposits are associated with many devastating diseases and fluorescent ligands able to visualize these pathological entities are essential. Here, we report the synthesis of thiophene‐based donor–acceptor–donor heptameric ligands that can be utilized for spectral assignment of distinct amyloid‐β (Aβ) aggregates, one of the pathological hallmarks in Alzheimer's disease. The ability of the ligands to selectively distinguish Aβ deposits was abolished when the chemical composition of the ligands was altered. Our findings provide the structural and functional basis for the development of new fluorescent ligands that can distinguish between aggregated proteinaceous species consisting of the same peptide or protein. In addition, such ligands might aid in interpreting the potential role of polymorphic Aβ deposits in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Linda Lantz
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Hamid Shirani
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Therése Klingstedt
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - K Peter R Nilsson
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| |
Collapse
|
45
|
New paradigms of clinical trial design for genetic prion diseases. Lancet Neurol 2020; 19:284-285. [DOI: 10.1016/s1474-4422(20)30029-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/03/2020] [Accepted: 01/17/2020] [Indexed: 11/23/2022]
|
46
|
Zakeri-Milani P, Shirani A, Nokhodchi A, Mussa Farkhani S, Mohammadi S, Shahbazi Mojarrad J, Mahmoudian M, Gholikhani T, Farshbaf M, Valizadeh H. Self-assembled peptide nanoparticles for efficient delivery of methotrexate into cancer cells. Drug Dev Ind Pharm 2020; 46:521-530. [PMID: 32116040 DOI: 10.1080/03639045.2020.1734017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The low cellular uptake of Methotrexate (MTX), a commonly used anticancer drug, is a big challenge for efficient cancer therapy. Self-assembled peptide nanoparticles (SAPNs) are one of the major classes of peptide vectors that have gained much attention toward novel drug delivery systems. In the present study, different sequences of cell-penetrating peptides including R2W4R2 and W3R4W3 and their SAPNs (R2W4R2-E12 and W3R4W3-E12) were designed for efficient delivery of MTX into MCF7 breast cancer cells. Based on electron microscopy results, the obtained SAPNs were in nano scale with spherical shape. There was a positive relationship between the free energy of water to octanol transferring and cellular penetration of designed nanostructures. The R2W4R2 possessed proper free energy and ability to form a spherical structure and hydrophobic-hydrophobic interactions, therefore, exhibited more cellular penetration than W3R4W3. The cellular uptake of obtained nanoparticles was examined by flow cytometry and fluorescence microscopy, in which, R2W4R2 and R2W4R2-E12 showed more appropriate penetration into MCF7 cells than W3R4W3 and W3R4W3-E12. The cytotoxicity of MTX-loaded peptides and SAPNs was examined by MTT assay. As a result, at higher concentrations, the R2W4R2 and R2W4R2-E12 showed higher cytotoxic behavior than their counterparts. Despite their enhanced cellular internalization, the cytotoxic behavior of MTX-loaded SAPNs at lower concentrations was relatively less than free MTX, which could be ascribed to the gradual nature of drug detachment from these conjugates. Therefore, R2W4R2 could be considered as an efficient choice to enhance the therapeutic efficiency of MTX in cancer treatments.
Collapse
Affiliation(s)
- Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Centre, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shirani
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Nokhodchi
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Falmer, Brighton, United Kingdom
| | - Samad Mussa Farkhani
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Samaneh Mohammadi
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Shahbazi Mojarrad
- Biotechnology Research Centre, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mahmoudian
- Drug Applied Research Centre, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tooba Gholikhani
- Biotechnology Research Centre, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Farshbaf
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Centre, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
47
|
Krance SH, Luke R, Shenouda M, Israwi AR, Colpitts SJ, Darwish L, Strauss M, Watts JC. Cellular models for discovering prion disease therapeutics: Progress and challenges. J Neurochem 2020; 153:150-172. [PMID: 31943194 DOI: 10.1111/jnc.14956] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
Prions, which cause fatal neurodegenerative disorders such as Creutzfeldt-Jakob disease, are misfolded and infectious protein aggregates. Currently, there are no treatments available to halt or even delay the progression of prion disease in the brain. The infectious nature of prions has resulted in animal paradigms that accurately recapitulate all aspects of prion disease, and these have proven to be instrumental for testing the efficacy of candidate therapeutics. Nonetheless, infection of cultured cells with prions provides a much more powerful system for identifying molecules capable of interfering with prion propagation. Certain lines of cultured cells can be chronically infected with various types of mouse prions, and these models have been used to unearth candidate anti-prion drugs that are at least partially efficacious when administered to prion-infected rodents. However, these studies have also revealed that not all types of prions are equal, and that drugs active against mouse prions are not necessarily effective against prions from other species. Despite some recent progress, the number of cellular models available for studying non-mouse prions remains limited. In particular, human prions have proven to be particularly challenging to propagate in cultured cells, which has severely hindered the discovery of drugs for Creutzfeldt-Jakob disease. In this review, we summarize the cellular models that are presently available for discovering and testing drugs capable of blocking the propagation of prions and highlight challenges that remain on the path towards developing therapies for prion disease.
Collapse
Affiliation(s)
- Saffire H Krance
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Russell Luke
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Marc Shenouda
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ahmad R Israwi
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Sarah J Colpitts
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Lina Darwish
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Maximilian Strauss
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
48
|
Caflisch A. Kinetic Control of Amyloidogenesis Calls for Unconventional Drugs To Fight Alzheimer's Disease. ACS Chem Neurosci 2020; 11:103-104. [PMID: 31904213 DOI: 10.1021/acschemneuro.9b00676] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Computer simulations had predicted that amyloid fibrillogenesis is governed by free energy barriers and kinetic traps (kinetic control), rather than the free energy of the final aggregates. The simulations suggested that the diversity in fibril morphologies can originate from variations in the number of protofilaments which has been confirmed by recent cryo-electron microscopy studies of amyloid-β fibrils derived from brain tissue of Alzheimer's patients. The kinetic control of fibril formation and polymorphism imply that chemical substances with new mechanisms of action are needed to fight Alzheimer's disease.
Collapse
Affiliation(s)
- Amedeo Caflisch
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| |
Collapse
|
49
|
Yuzu K, Lindgren M, Nyström S, Zhang J, Mori W, Kunitomi R, Nagase T, Iwaya K, Hammarström P, Zako T. Insulin amyloid polymorphs: implications for iatrogenic cytotoxicity. RSC Adv 2020; 10:37721-37727. [PMID: 35515176 PMCID: PMC9057202 DOI: 10.1039/d0ra07742a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Amyloid specific fluorescent probes are becoming an important tool for studies of disease progression and conformational polymorphisms in diseases related to protein misfolding and aggregation such as localized and systemic amyloidosis. Herein, it is demonstrated that using the amyloid specific fluorescent probes pFTAA and benzostyryl capped benzothiadiazole BTD21, structural polymorphisms of insulin amyloids are imaged in localized insulin-derived amyloid aggregates formed at subcutaneous insulin-injection sites in patients with diabetes. It is also found that pFTAA and BTD21 could discriminate structural polymorphisms of insulin amyloids, so called fibrils and filaments, formed in vitro. In addition, it is shown that insulin drug preparations used for treating diabetes formed various types of amyloid aggregates that can be assessed and quantified using pFTAA and BTD21. Interestingly, incubated pFTAA-positive insulin preparation aggregates show cytotoxicity while BTD21-positive aggregates are less toxic. From these observations, a variety of amyloid polymorphic structures with different cytotoxicities formed both in vivo and in vitro by various insulin preparations are proposed. Structural polymorphism of insulin amyloids in vivo can be recognized using novel amyloid specific fluorescent probes, pFTAA and BTD21.![]()
Collapse
Affiliation(s)
- Keisuke Yuzu
- Department of Chemistry and Biology
- Graduate School of Science and Engineering
- Ehime University
- Matsuyama
- Japan
| | - Mikael Lindgren
- Department of Physics
- Faculty of Natural Sciences
- Norwegian University of Science and Technology
- NO-7491 Trondheim
- Norway
| | - Sofie Nyström
- IFM Chemistry
- Linköping University
- SE-58183 Linköping
- Sweden
| | - Jun Zhang
- IFM Chemistry
- Linköping University
- SE-58183 Linköping
- Sweden
| | - Wakako Mori
- Department of Chemistry and Biology
- Graduate School of Science and Engineering
- Ehime University
- Matsuyama
- Japan
| | - Risako Kunitomi
- Department of Chemistry and Biology
- Graduate School of Science and Engineering
- Ehime University
- Matsuyama
- Japan
| | - Terumasa Nagase
- Department of Metabolism and Endocrinology
- Tokyo Medical University Ibaraki Medical Center
- Japan
| | - Keiichi Iwaya
- Department of Pathology
- Sasaki Institute
- Kyoundo Hospital
- Tokyo 101-0062
- Japan
| | | | - Tamotsu Zako
- Department of Chemistry and Biology
- Graduate School of Science and Engineering
- Ehime University
- Matsuyama
- Japan
| |
Collapse
|
50
|
Holec SA, Block AJ, Bartz JC. The role of prion strain diversity in the development of successful therapeutic treatments. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 175:77-119. [PMID: 32958242 PMCID: PMC8939712 DOI: 10.1016/bs.pmbts.2020.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Prions are a self-propagating misfolded conformation of a cellular protein. Prions are found in several eukaryotic organisms with mammalian prion diseases encompassing a wide range of disorders. The first recognized prion disease, the transmissible spongiform encephalopathies (TSEs), affect several species including humans. Alzheimer's disease, synucleinopathies, and tauopathies share a similar mechanism of self-propagation of the prion form of the disease-specific protein reminiscent of the infection process of TSEs. Strain diversity in prion disease is characterized by differences in the phenotype of disease that is hypothesized to be encoded by strain-specific conformations of the prion form of the disease-specific protein. Prion therapeutics that target the prion form of the disease-specific protein can lead to the emergence of drug-resistant strains of prions, consistent with the hypothesis that prion strains exist as a dynamic mixture of a dominant strain in combination with minor substrains. To overcome this obstacle, therapies that reduce or eliminate the template of conversion are efficacious, may reverse neuropathology, and do not result in the emergence of drug resistance. Recent advancements in preclinical diagnosis of prion infection may allow for a combinational approach that treats the prion form and the precursor protein to effectively treat prion diseases.
Collapse
Affiliation(s)
- Sara A.M. Holec
- Institute for Applied Life Sciences and Department of Biology, University of Massachusetts Amherst, Amherst, MA, United States,Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, United States
| | - Alyssa J. Block
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, United States
| | - Jason C. Bartz
- Department of Medical Microbiology and Immunology, School of Medicine, Creighton University, Omaha, NE, United States,Corresponding author:
| |
Collapse
|