1
|
Roy S, Biswas S, Nandy A, Guha D, Dasgupta R, Bagchi A, Sil PC. An approach to predict and inhibit Amyloid Beta dimerization pattern in Alzheimer’s disease. Toxicol Rep 2025; 14:101879. [DOI: 10.1016/j.toxrep.2024.101879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
|
2
|
Ahmad S, Gardner QA, Shakir NA, Gulzar S, Azim N, Akhtar M. Nature of recombinant human serum amyloid A1 in Escherichia coli and its preferable approach for purification. Protein Expr Purif 2025; 227:106620. [PMID: 39505093 DOI: 10.1016/j.pep.2024.106620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024]
Abstract
Serum amyloid A1 (SAA1) is an apolipoprotein which is involved in amyloid A amyloidosis (AA) by forming fibrils. The process of fibrillation is still being explored and holds challenges in recombinant expression and purification of SAA1. This study deals with the preferable approach for the expression and purification of SAA1 which is normally toxic and unstable to express without using any fusion-tag. Complete soluble expression of SAA1 was obtained without the use of additional tag, in terrific broth, supplemented with 3 % ethanol at 30 °C. Soluble fraction of SAA1 was initially treated with salting-out using ammonium sulphate giving 1.5 M salt concentration to avoid SAA1 protein precipitation along with unwanted proteins. The soluble fraction of SAA1 after salting-out was purified by two individual chromatographic approaches: One anion exchange and second reverse phase chromatography. The yield of purified SAA1 was 3 times greater by anion exchange than reverse phase chromatography. MALDI-TOF analysis of purified SAA1 showed 11813 Da for intact protein and proteome analysis revealed greater than 90 % sequence coverage by MASCOT. The subunit interaction showed hexamer form at basic pH which was analyzed by size exclusion chromatography. The fibrillation activity of SAA1 was found to be 10-15 times higher in basic media at 43 °C than 37 °C. Our research demonstrates successful expression and purification of wild-type human recombinant SAA1. The cost-effective radical approach employed for purification of SAA1 is crucial for thorough protein characterization particularly, mechanisms of protein aggregation involved in amyloidosis.
Collapse
Affiliation(s)
- Saira Ahmad
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore, 54590, Pakistan
| | - Qurratulann Afza Gardner
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore, 54590, Pakistan.
| | - Nisar Ahmad Shakir
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore, 54590, Pakistan
| | - Sabahat Gulzar
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore, 54590, Pakistan
| | - Naseema Azim
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore, 54590, Pakistan
| | - Muhammad Akhtar
- School of Biological Sciences, University of the Punjab, Quaid-e-Azam Campus, Lahore, 54590, Pakistan; Biological Sciences, University of Southampton SO17 1BJ, UK
| |
Collapse
|
3
|
Wang Y, Gao P, Wu Z, Jiang B, Wang Y, He Z, Zhao B, Tian X, Gao H, Cai L, Li W. Exploring the therapeutic potential of Chinese herbs on comorbid type 2 diabetes mellitus and Parkinson's disease: A mechanistic study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119095. [PMID: 39537117 DOI: 10.1016/j.jep.2024.119095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/12/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Type 2 diabetes mellitus (T2DM) and Parkinson's disease (PD) are chronic conditions that affect the aging population, with increasing prevalence globally. The rising prevalence of comorbidity between these conditions, driven by demographic shifts, severely impacts the quality of life of patients, posing a significant burden on healthcare resources. Chinese herbal medicine has been used to treat T2DM and PD for millennia. Pharmacological studies have demonstrated that medicinal herbs effectively lower blood glucose levels and exert neuroprotective effects, suggesting their potential as adjunctive therapy for concurrent management of T2DM and PD. AIM OF THE STUDY To elucidate the shared mechanisms underlying T2DM and PD, particularly focusing on the potential mechanisms by which medicinal herbs (including herbal formulas, single herbs, and active compounds) may treat these diseases, to provide valuable insights for developing therapeutics targeting comorbid T2DM and PD. MATERIALS AND METHODS Studies exploring the mechanisms underlying T2DM and PD, as well as the treatment of these conditions with medicinal herbs, were extracted from several electronic databases, including PubMed, Web of Science, Google Scholar, and China National Knowledge Infrastructure (CNKI). RESULTS Numerous studies have shown that inflammation, oxidative stress, insulin resistance, impaired autophagy, gut microbiota dysbiosis, and ferroptosis are shared mechanisms underlying T2DM and PD mediated through the NLRP3 inflammasome, NF-κB, MAPK, Keap1/Nrf2/ARE, PI3K/AKT, AMPK/SIRT1, and System XC--GSH-GPX4 signaling pathways. Thirty-four medicinal herbs, including 2 herbal formulas, 4 single herbs, and 28 active compounds, have been reported to potentially exert anti-T2DM and anti-PD effects by targeting these shared mechanisms. CONCLUSIONS Traditional Chinese medicine effectively combats T2DM and PD through shared pathological mechanisms, highlighting their potential for application in treating these comorbid conditions.
Collapse
Affiliation(s)
- Yan Wang
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Pengpeng Gao
- Department of Preventive Treatment, Ningxia Integrated Chinese and Western Medicine Hospital, Yinchuan, 750004, China
| | - Zicong Wu
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yanru Wang
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Zhaxicao He
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Bing Zhao
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xinyun Tian
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Han Gao
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Li Cai
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Wentao Li
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
4
|
Sakunthala A, Maji SK. Deciphering the Seed Size-Dependent Cellular Internalization Mechanism for α-Synuclein Fibrils. Biochemistry 2025; 64:377-400. [PMID: 39762762 DOI: 10.1021/acs.biochem.4c00667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Aggregation of α-synuclein (α-Syn) and Lewy body (LB) formation are the key pathological events implicated in Parkinson's disease (PD) that spread in a prion-like manner. However, biophysical and structural characteristics of toxic α-Syn species and molecular events that drive early events in the propagation of α-Syn amyloids in a prion-like manner remain elusive. We used a neuronal cell model to demonstrate the size-dependent native biological activities of α-Syn fibril seeds. Biophysical characterization of the fibril seeds generated by controlled fragmentation indicated that increased fragmentation leads to a reduction in fibril size, correlating directly with the extent of fragmentation events. Although the size-based complexity of amyloid fibrils modulates their biological activities and fibril amplification pathways, it remains unclear how the variability of fibril seed size dictates its specific uptake mechanism into the cells. The present study elucidates the mechanism of α-Syn fibril internalization and how it is regulated by the size of fibril seeds. Further, we demonstrate that size-dependent endocytic pathways (dynamin-dependent clathrin/caveolin-mediated) are more prominent for the differential uptake of short fibril seeds compared to their longer counterparts. This size-dependent preference might contribute to the enhanced uptake and transcellular propagation of short α-Syn fibril seeds in a prion-like manner. Overall, the present study suggests that the physical dimension of α-Syn amyloid fibril seeds significantly influences their cellular uptake and pathological responses in the initiation and progression of PD.
Collapse
Affiliation(s)
- Arunima Sakunthala
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases (SCAN), Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
- Department of Biosciences& Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Samir K Maji
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases (SCAN), Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
- Department of Biosciences& Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
5
|
Larsen JA, Barclay A, Vettore N, Klausen LK, Mangels LN, Coden A, Schmit JD, Lindorff-Larsen K, Buell AK. The mechanism of amyloid fibril growth from Φ-value analysis. Nat Chem 2025:10.1038/s41557-024-01712-9. [PMID: 39820805 DOI: 10.1038/s41557-024-01712-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/29/2024] [Indexed: 01/19/2025]
Abstract
Amyloid fibrils are highly stable misfolded protein assemblies that play an important role in several neurodegenerative and systemic diseases. Although structural information of the amyloid state is now abundant, mechanistic details about the misfolding process remain elusive. Inspired by the Φ-value analysis of protein folding, we combined experiments and molecular simulations to resolve amino-acid contacts and determine the structure of the transition-state ensemble-the rate-limiting step-for fibril elongation of PI3K-SH3 amyloid fibrils. The ensemble was validated experimentally by Tanford β analysis and computationally by free energy calculations. Although protein folding proceeds on funnel-shaped landscapes, here we find that the energy landscape for the misfolding reaction consists of a large 'golf course' region, defined by a single energy barrier and transition state, accessing a sharply funnelled region. Thus, misfolding occurs by rare, successful monomer-fibril end collisions interspersed by numerous unsuccessful binding attempts. Taken together, these insights provide a quantitative and highly resolved description of a protein misfolding reaction.
Collapse
Affiliation(s)
- Jacob Aunstrup Larsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Abigail Barclay
- Structural Biophysics, Niels Bohr Institute, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Structural Biology and NMR Laboratory & the Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Nicola Vettore
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Louise K Klausen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Lena N Mangels
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Alberto Coden
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Jeremy D Schmit
- Department of Physics, Kansas State University, Manhattan, KS, USA
| | - Kresten Lindorff-Larsen
- Structural Biology and NMR Laboratory & the Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Alexander K Buell
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark.
| |
Collapse
|
6
|
Gao Z, Jing B, Wang Y, Wan W, Dong X, Liu Y. Exploring the impact of lipid nanoparticles on protein stability and cellular proteostasis. J Colloid Interface Sci 2025; 678:656-665. [PMID: 39216393 DOI: 10.1016/j.jcis.2024.08.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Lipid nanoparticles (LNPs) have become pivotal in advancing modern medicine, from mRNA-based vaccines to gene editing with CRISPR-Cas9 systems. Though LNPs based therapeutics offer promising drug delivery with satisfactory clinical safety profiles, concerns are raised regarding their potential nanotoxicity. Here, we explore the impacts of LNPs on protein stability in buffer and cellular protein homeostasis (proteostasis) in HepG2 cells. First, we show that LNPs of different polyethylene glycol (PEG) molar ratios to total lipid ratio boost protein aggregation propensity by reducing protein stability in cell lysate and blood plasma. Second, in HepG2 liver cells, these LNPs induce global proteome aggregation, as imaged by a cellular protein aggregation fluorescent dye (AggStain). Such LNPs induced proteome aggregation is accompanied by decrease in cellular micro-environmental polarity as quantified by a solvatochromic protein aggregation sensor (AggRetina). The observed local polarity fluctuations may be caused by the hydrophobic contents of LNPs that promote cellular proteome aggregation. Finally, we exploit RNA sequencing analysis (RNA-Seq) to reveal activation of unfolded protein response (UPR) pathway and other proteostasis genes upon LNPs treatment. Together, these findings highlight that LNPs may induce subtle proteome stress by compromising protein stability and proteostasis even without obvious damage to cell viability.
Collapse
Affiliation(s)
- Zifan Gao
- Dalian Medical University, Dalian 116044, China
| | - Biao Jing
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Yuhui Wang
- The Second Hospital of Dalian Medical University, Dalian 116023, China
| | - Wang Wan
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| | - Xuepeng Dong
- The Second Hospital of Dalian Medical University, Dalian 116023, China.
| | - Yu Liu
- State Key Laboratory of Medical Proteomics, National Chromatographic R. & A. Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China.
| |
Collapse
|
7
|
Davoodi S, Ornithopoulou E, Gavillet CJ, Davydok A, Roth SV, Lendel C, Lundell F. Confinement-Induced Self-Assembly of Protein Nanofibrils Probed by Microfocus X-ray Scattering. J Phys Chem B 2025. [PMID: 39808180 DOI: 10.1021/acs.jpcb.4c04386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
We here explore confinement-induced assembly of whey protein nanofibrils (PNFs) into microscale fibers using microfocused synchrotron X-ray scattering. Solvent evaporation aligns the PNFs into anisotropic fibers, and the process is followed in situ by scattering experiments within a droplet of PNF dispersion. We find an optimal temperature at which the order parameter of the protein fiber is maximized, suggesting that the degree of order results from a balance between the time scales of the forced alignment and the rotational diffusion of the fibrils. Furthermore, the assembly process is shown to depend on the nanoscale morphology and flexibility of the PNFs. Stiff/straight PNFs with long persistence lengths (∼2 μm) align at the air-water interface, with anisotropy decreasing toward the center of the droplet as Marangoni flows increase entanglement toward the center. By contrast, flexible/curved PNFs with shorter persistence lengths (<100 nm) align more uniformly throughout the droplet, likely due to enhanced local entanglements. Straight PNFs pack tightly, forming smaller clusters with short intercluster distances, while curved PNFs form intricate, adaptable networks with larger characteristic distances and more varied structures.
Collapse
Affiliation(s)
- Saeed Davoodi
- Department of Engineering Mechanics, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
- Wallenberg Wood Science Center, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| | - Eirini Ornithopoulou
- Department of Engineering Mechanics, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
- Department of Chemistry, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| | - Calvin J Gavillet
- Department of Engineering Mechanics, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
- Wallenberg Wood Science Center, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
- Deutsches Elektronen-Synchrotron, D-22607 Hamburg, Germany
| | - Anton Davydok
- Institute of Materials Research, Helmholtz-Zentrum Geesthacht, D-22607 Hamburg, Germany
| | - Stephan V Roth
- Wallenberg Wood Science Center, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
- Deutsches Elektronen-Synchrotron, D-22607 Hamburg, Germany
- Department of Fibre and Polymer Technology, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| | - Christofer Lendel
- Department of Chemistry, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| | - Fredrik Lundell
- Department of Engineering Mechanics, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
- Wallenberg Wood Science Center, KTH Royal Institute of Technology, 100 44 Stockholm, Sweden
| |
Collapse
|
8
|
Haghparas Z, Bouloorchi Tabalvandani M, Arghavani P, Behjati Hosseini S, Badieirostami M, Habibi-Rezaei M, Moosavi-Movahedi AA. Modeling the navigating forces behind BSA aggregation in a microfluidic chip. SOFT MATTER 2025. [PMID: 39807901 DOI: 10.1039/d4sm01148a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Microfluidic chips are powerful tools for investigating numerous variables including chemical and physical parameters on protein aggregation. This study investigated the aggregation of bovine serum albumin (BSA) in two different systems: a vial-based static system and a microfluidic chip-based dynamic system in which BSA aggregation was induced successfully. BSA aggregation induced in a microfluidic chip on a timescale of seconds enabled a dynamic investigation of the forces driving the aggregation process. This study employed a combination of experimental approaches, including biophysical and microscopic methods, and computational simulations using MATLAB and COMSOL Multiphysics. Obtained results revealed that Brownian movement, advective mixing, and laminar flow applied in favor of the formation of amyloid-like aggregates through the entire pathway. Furthermore, heating provided the necessary energy for the initial BSA's partial unfolding. In the following, space restriction and the cumulative effects of repulsive electrostatic and attractive van der Waals forces contributed to forming BSA clusters as a partially unfolded intermediate in the first few seconds of the aggregation process. Consequently, the synergistic effects of hydrodynamic forces (including shear force), hydrophobic interaction, and space restriction resulted in the deposition of larger aggregates on the channel sidewalls. Due to the elevated local concentration of BSA clusters alongside the strong shear force toward the channel sidewalls, the deposited structures underwent a structural conversion to form amyloid-like aggregates within a few seconds. In this study, we not only elucidated the molecular mechanisms underlying BSA aggregation but also highlighted the forces driving the aggregation process in microfluidic systems, explaining how it occurs within a timescale of seconds.
Collapse
Affiliation(s)
- Zahra Haghparas
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | | | - Payam Arghavani
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | | | - Majid Badieirostami
- MEMS Lab, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | | | | |
Collapse
|
9
|
Yang L, Zhang J, Andon JS, Li L, Wang T. Rapid discovery of cyclic peptide protein aggregation inhibitors by continuous selection. Nat Chem Biol 2025:10.1038/s41589-024-01823-x. [PMID: 39806068 DOI: 10.1038/s41589-024-01823-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
Protein aggregates are associated with numerous diseases. Here we report a platform for the rapid phenotypic selection of protein aggregation inhibitors from genetically encoded cyclic peptide libraries in Escherichia coli based on phage-assisted continuous evolution (PACE). We developed a new PACE-compatible selection for protein aggregation inhibition and used it to identify cyclic peptides that suppress amyloid-β42 and human islet amyloid polypeptide aggregation. Additionally, we integrated a negative selection that removes false positives and off-target hits, greatly improving cyclic peptide selectivity. We show that selected inhibitors are active when chemically resynthesized in in vitro assays. Our platform provides a powerful approach for the rapid discovery of cyclic peptide inhibitors of protein aggregation and may serve as the basis for the future evolution of cyclic peptides with a broad spectrum of inhibitory activities.
Collapse
Affiliation(s)
- Linwei Yang
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jingwei Zhang
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - James S Andon
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Tina Wang
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
10
|
Urbanek A, Garland EF, Prescott EE, King MC, Olerinyova A, Wareing HE, Georgieva N, Bradshaw EL, Tzokov SB, Knight A, Tartakovskii AI, Malm T, Highley JR, De S. Molecular Determinants of Protein Pathogenicity at the Single-Aggregate Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2410229. [PMID: 39804980 DOI: 10.1002/advs.202410229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Determining the structure-function relationships of protein aggregates is a fundamental challenge in biology. These aggregates, whether formed in vitro, within cells, or in living organisms, present significant heterogeneity in their molecular features such as size, structure, and composition, making it difficult to determine how their structure influences their functions. Interpreting how these molecular features translate into functional roles is crucial for understanding cellular homeostasis and the pathogenesis of various debilitating diseases like Alzheimer's and Parkinson's. In this study, a bottom-up approach is introduced to explore how variations in protein aggregates' size, composition, post-translational modifications and point mutations profoundly influence their biological functions. Applying this method to Alzheimer's and Parkinson's associated proteins, novel disease-relevant pathways are uncovered, demonstrating how subtle alterations in composition and morphology can shift the balance between healthy and pathological states. This findings provide deeper insights into the molecular basis of protein's functions at the single-aggregate level, enhancing the knowledge of their roles in health and disease.
Collapse
Affiliation(s)
- Agnieszka Urbanek
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Emma F Garland
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Emily E Prescott
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Marianne C King
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Anna Olerinyova
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Hollie E Wareing
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Nia Georgieva
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Ellie L Bradshaw
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Svetomir B Tzokov
- Cryo-Electron Microscopy Facility, School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| | - Alexander Knight
- Department of Physics and Astronomy, University of Sheffield, Sheffield, S3 7RH, UK
| | | | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, 70211, Finland
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Suman De
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| |
Collapse
|
11
|
Horvath I, Aning OA, Kk S, Rehnberg N, Chawla S, Molin M, Westerlund F, Wittung-Stafshede P. Biological Amyloids Chemically Damage DNA. ACS Chem Neurosci 2025. [PMID: 39782739 DOI: 10.1021/acschemneuro.4c00461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
Amyloid fibrils are protein polymers noncovalently assembled through β-strands arranged in a cross-β structure. Biological amyloids were considered chemically inert until we and others recently demonstrated their ability to catalyze chemical reactions in vitro. To further explore the functional repertoire of amyloids, we here probe if fibrils of α-synuclein (αS) display chemical reactivity toward DNA. We demonstrate that αS amyloids bind DNA at micromolar concentrations in vitro. Using the activity of DNA repair enzymes as proxy for damage, we unravel that DNA-amyloid interactions promote chemical modifications, such as single-strand nicks, to the DNA. Double-strand breaks are also evident based on nanochannel analysis of individual long DNA molecules. The amyloid fold is essential for the activity as no DNA chemical modification is detected with αS monomers. In a yeast cell model, there is increased DNA damage when αS is overexpressed. Chemical perturbation of DNA adds another chemical reaction to the set of activities emerging for biological amyloids. Since αS amyloids are also found in the nuclei of neuronal cells of Parkinson's disease (PD) patients, and increased DNA damage is a hallmark of PD, we propose that αS amyloids contribute to PD by direct chemical perturbation of DNA.
Collapse
Affiliation(s)
- Istvan Horvath
- Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Obed Akwasi Aning
- Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Sriram Kk
- Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Nikita Rehnberg
- Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Srishti Chawla
- Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Mikael Molin
- Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | - Fredrik Westerlund
- Department of Life Sciences, Chalmers University of Technology, 412 96 Gothenburg, Sweden
| | | |
Collapse
|
12
|
Mancl JM, Liang WG, Bayhi NL, Wei H, Carragher B, Potter CS, Tang WJ. Characterization and modulation of human insulin degrading enzyme conformational dynamics to control enzyme activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.30.630732. [PMID: 39803525 PMCID: PMC11722313 DOI: 10.1101/2024.12.30.630732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Insulin degrading enzyme (IDE) is a dimeric 110 kDa M16A zinc metalloprotease that degrades amyloidogenic peptides diverse in shape and sequence, including insulin, amylin, and amyloid-β, to prevent toxic amyloid fibril formation. IDE has a hollow catalytic chamber formed by four homologous subdomains organized into two ~55 kDa N- and C- domains (IDE-N and IDE-C, respectively), in which peptides bind, unfold, and are repositioned for proteolysis. IDE is known to transition between a closed state, poised for catalysis, and an open state, able to release cleavage products and bind new substrate. Here, we present five cryoEM structures of the IDE dimer at 3.0-4.1 Å resolution, obtained in the presence of a sub-saturating concentration of insulin. Analysis of the heterogeneity within the particle populations comprising these structures combined with all-atom molecular dynamics (MD) simulations permitted a comprehensive characterization of IDE conformational dynamics. Our analysis identified the structural basis and key residues for these dynamics that were not revealed by IDE static structures. Notably arginine-668 serves as a molecular latch mediating the open-close transition and facilitates key protein motions through charge-swapping interactions at the IDE-N/C interface. Our size-exclusion chromatography-coupled small-angle X-ray scattering and enzymatic assays of an arginine-668 to alanine mutant indicate a profound alteration of conformational dynamics and catalytic activity. Taken together, this work highlights the power of integrating experimental and computational methodologies to understand protein dynamics, offers the molecular basis of unfoldase activity of IDE, and provides a new path forward towards the development of substrate-specific modulators of IDE activity.
Collapse
Affiliation(s)
- Jordan M. Mancl
- Ben-May Institute for Cancer Research, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, USA
| | - Wenguang G. Liang
- Ben-May Institute for Cancer Research, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, USA
- Simons Electron Microscopy Center, New York Structural Biology Center, 89 Convent Ave, New York, New York, 10027, USA
| | - Nicholas L. Bayhi
- Biophysics Science Graduate Program, The University of Chicago, 929 East 57th street, Chicago, Illinois 60637, USA
| | - Hui Wei
- Simons Electron Microscopy Center, New York Structural Biology Center, 89 Convent Ave, New York, New York, 10027, USA
| | - Bridget Carragher
- Simons Electron Microscopy Center, New York Structural Biology Center, 89 Convent Ave, New York, New York, 10027, USA
- Current affiliation - Chan Zuckerberg Institute for Advanced Biological Imaging
| | - Clinton S. Potter
- Simons Electron Microscopy Center, New York Structural Biology Center, 89 Convent Ave, New York, New York, 10027, USA
- Current affiliation - Chan Zuckerberg Institute for Advanced Biological Imaging
| | - Wei-Jen Tang
- Ben-May Institute for Cancer Research, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, USA
| |
Collapse
|
13
|
Zhang S, Owyong TC, Sanislav O, Englmaier L, Sui X, Wang G, Greening DW, Williamson NA, Villunger A, White JM, Heras B, Wong WWH, Fisher PR, Hong Y. Global analysis of endogenous protein disorder in cells. Nat Methods 2025; 22:124-134. [PMID: 39587358 DOI: 10.1038/s41592-024-02507-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 10/14/2024] [Indexed: 11/27/2024]
Abstract
Disorder and flexibility in protein structures are essential for biological function but can also contribute to diseases, such as neurodegenerative disorders. However, characterizing protein folding on a proteome-wide scale within biological matrices remains challenging. Here we present a method using a bifunctional chemical probe, named TME, to capture in situ, enrich and quantify endogenous protein disorder in cells. TME exhibits a fluorescence turn-on effect upon selective conjugation with proteins with free cysteines in surface-exposed and flexible environments-a distinctive signature of protein disorder. Using an affinity-based proteomic approach, we identify both basal disordered proteins and those whose folding status changes under stress, with coverage to proteins even of low abundance. In lymphoblastoid cells from individuals with Parkinson's disease and healthy controls, our TME-based strategy distinguishes the two groups more effectively than lysate profiling methods. High-throughput TME fluorescence and proteomics further reveal a universal cellular quality-control mechanism in which cells adapt to proteostatic stress by adopting aggregation-prone distributions and sequestering disordered proteins, as illustrated in Huntington's disease cell models.
Collapse
Affiliation(s)
- Shouxiang Zhang
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Tze Cin Owyong
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Oana Sanislav
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Lukas Englmaier
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Xiaojing Sui
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, IL, USA
| | - Geqing Wang
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Melbourne, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia
| | - Nicholas A Williamson
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Villunger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute for Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Jonathan M White
- School of Chemistry, Bio21 Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Begoña Heras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Wallace W H Wong
- School of Chemistry, Bio21 Institute, The University of Melbourne, Parkville, Victoria, Australia
- ARC Centre of Excellence in Exciton Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul R Fisher
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Yuning Hong
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
14
|
Mitra A, Chakraborty D, Naik L, Dhiman R, Sarkar N. Anti-amyloidogenic hexapeptide-coated gold nanoparticles for enhanced inhibition of amyloid formation: A promising therapeutic approach. Int J Biol Macromol 2025; 284:138002. [PMID: 39586437 DOI: 10.1016/j.ijbiomac.2024.138002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
Under specific external stimulus, misfolded and natively disordered globular proteins undergo irreversible transformation into pathogenic β-sheet-rich insoluble fibrillar structure, and deposition of theses fibrils in cells and tissues leads to disorders like Alzheimer's, Dementia, Type II diabetes, and many more. Here, we have developed a positively-charged Arg-containing hexapeptide, SqP7, and elucidated its anti-amyloidogenic propensity on in vitro HEWL amyloid formation under acidic and neutral fibrillation conditions using computational tools and several biophysical techniques. SqP7, at a five-fold molar excess, displayed excellent amyloid inhibition capability at both pH conditions (~83 % and 72 % inhibition under acidic and neutral fibrillation conditions, respectively), and was further chosen as a coating agent on gold nanoparticles. This was done to investigate whether coating of this peptide on gold nanoparticles has any effect on its anti-amyloidogenic efficiency and effective inhibition concentration. The synthesized SqP7-coated gold nanoparticles were characterized to be spherical and highly-dispersed having a mean diameter of 9.12 ± 2.08 nm. The anti-amyloidogenic capability of the synthesized SqP7-coated gold nanoparticles was further evaluated, and a 10-fold reduction in the effective inhibition concentration of SqP7 was observed. This peptide‑gold nanoparticle based integrated approach can lead to the development of highly effective therapeutics for amyloid-related diseases, offering improved prevention and treatment options.
Collapse
Affiliation(s)
- Amit Mitra
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Debashmita Chakraborty
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Lincoln Naik
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India
| | - Nandini Sarkar
- Department of Biotechnology and Medical Engineering, National Institute of Technology Rourkela, Rourkela- 769008, Odisha, India.
| |
Collapse
|
15
|
Spivak I, Guldiken N, Usachov V, Schaap F, Damink SWO, Bouchecareilh M, Lehmann A, Fu L, Mo F, Ensari GK, Hufnagel F, Fromme M, Preisinger C, Strnad P. Alpha-1 Antitrypsin Inclusions Sequester GRP78 in a Bile Acid-Inducible Manner. Liver Int 2025; 45:e16207. [PMID: 39665869 PMCID: PMC11636636 DOI: 10.1111/liv.16207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND AND AIMS The homozygous PiZ mutation (PIZZ genotype) constitutes the predominant cause of severe alpha-1 antitrypsin (AAT) deficiency and leads to liver disease via hepatocellular AAT aggregation. We systematically analysed the composition of AAT aggregates and studied the impact of bile acids. METHODS AAT inclusions were isolated from livers of PiZ overexpressing mice and PIZZ humans via fluorescence-activated and immunomagnetic sorting (FACS/MACS), while insoluble proteins were obtained via Triton-X extraction. Inclusion composition was evaluated through mass-spectrometry (MS), immunoblotting and immunostaining. Hepatocytes with versus without AAT aggregates were obtained via microdissection. Serum bile acids were assessed in 57 PIZZ subjects and 19 controls. Mice were administered 2% cholic acid (CA)-supplemented chow for 7 days. RESULTS MS identified the key endoplasmic reticulum chaperone 78 kDa glucose-regulated protein (GRP78) in FACS/MACS pulldowns. GRP78 was also enriched in insoluble fractions from PiZ mice versus wild types and detected in insoluble fractions/MACS isolates from PIZZ liver explants. In cultured cells/primary hepatocytes, PiZ overexpression was associated with increased GRP78 mRNA/protein levels. In human livers, hepatocytes with AAT aggregates had higher GRP78 levels than hepatocytes without. PIZZ subjects displayed higher serum bile acid levels than controls and the highest levels were seen in individuals with liver injury/fibrosis. In PiZ mice, CA-mediated bile acid challenge resulted in increased liver injury and translocation of GRP78 into the aggregates. CONCLUSIONS Our results demonstrate that GRP78 is sequestered within AAT inclusions. Bile acid accumulation, as seen in PIZZ subjects with liver disease, may promote GRP78 segregation and thereby augment liver damage. TRIAL REGISTRATION NCT02929940.
Collapse
Affiliation(s)
- Igor Spivak
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Nurdan Guldiken
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Valentyn Usachov
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Frank Schaap
- Department of Surgery, Maastricht University Medical Center and NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtNetherlands
- Department of General, Visceral and Transplant SurgeryUniversity Hospital RWTH AachenAachenGermany
| | - Steven W.M. Olde Damink
- Department of Surgery, Maastricht University Medical Center and NUTRIM School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtNetherlands
- Department of General, Visceral and Transplant SurgeryUniversity Hospital RWTH AachenAachenGermany
| | | | | | - Lei Fu
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
- Department of Science and TechnologyRuikang Hospital Affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Fa‐Rong Mo
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Gökce Kobazi Ensari
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Franziska Hufnagel
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Malin Fromme
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Christian Preisinger
- Interdisciplinary Center for Clinical Research (IZKF)University Hospital RWTH AachenAachenGermany
| | - Pavel Strnad
- Medical Department III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| |
Collapse
|
16
|
Pintado-Grima C, Ventura S. The role of amphipathic and cationic helical peptides in Parkinson's disease. Protein Sci 2025; 34:e70020. [PMID: 39720890 DOI: 10.1002/pro.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/29/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024]
Abstract
Peptides are attracting a growing interest for therapeutic applications in biomedicine. In Parkinson's disease (PD), different human endogenous peptides have been associated with beneficial effects, including protein aggregation inhibition, reduced inflammation, or the protection of dopaminergic neurons. Such effects seem to be connected to the spatial arrangement of peptide side chains, and many of these human molecules share common conformational traits, displaying a distinctive amphipathic and cationic helical structure, which is believed to be crucial for their activities. This review delves into the relationship between these structural properties and the current evidence connecting biogenic peptides to the amelioration of PD symptoms. We discuss their implications in the disease, the different mechanisms of action, their state of validation, and their therapeutic potential.
Collapse
Affiliation(s)
- Carlos Pintado-Grima
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
- Hospital Universitari Parc Taulí, Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Sabadell, Spain
| |
Collapse
|
17
|
Vassallo N. Poration of mitochondrial membranes by amyloidogenic peptides and other biological toxins. J Neurochem 2025; 169:e16213. [PMID: 39213385 DOI: 10.1111/jnc.16213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria are essential organelles known to serve broad functions, including in cellular metabolism, calcium buffering, signaling pathways and the regulation of apoptotic cell death. Maintaining the integrity of the outer (OMM) and inner mitochondrial membranes (IMM) is vital for mitochondrial health. Cardiolipin (CL), a unique dimeric glycerophospholipid, is the signature lipid of energy-converting membranes. It plays a significant role in maintaining mitochondrial architecture and function, stabilizing protein complexes and facilitating efficient oxidative phosphorylation (OXPHOS) whilst regulating cytochrome c release from mitochondria. CL is especially enriched in the IMM and at sites of contact between the OMM and IMM. Disorders of protein misfolding, such as Alzheimer's and Parkinson's diseases, involve amyloidogenic peptides like amyloid-β, tau and α-synuclein, which form metastable toxic oligomeric species that interact with biological membranes. Electrophysiological studies have shown that these oligomers form ion-conducting nanopores in membranes mimicking the IMM's phospholipid composition. Poration of mitochondrial membranes disrupts the ionic balance, causing osmotic swelling, loss of the voltage potential across the IMM, release of pro-apoptogenic factors, and leads to cell death. The interaction between CL and amyloid oligomers appears to favour their membrane insertion and pore formation, directly implicating CL in amyloid toxicity. Additionally, pore formation in mitochondrial membranes is not limited to amyloid proteins and peptides; other biological peptides, as diverse as the pro-apoptotic Bcl-2 family members, gasdermin proteins, cobra venom cardiotoxins and bacterial pathogenic toxins, have all been described to punch holes in mitochondria, contributing to cell death processes. Collectively, these findings underscore the vulnerability of mitochondria and the involvement of CL in various pathogenic mechanisms, emphasizing the need for further research on targeting CL-amyloid interactions to mitigate mitochondrial dysfunction.
Collapse
Affiliation(s)
- Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Tal-Qroqq, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Tal-Qroqq, Malta
| |
Collapse
|
18
|
Batys P, Krzemień L, Barbasz J. dRama: Differential Ramachandran Plot as a Tool to Analyze Subtle Changes in Protein Secondary Structure. Proteomics Clin Appl 2025; 19:e202400087. [PMID: 39582098 DOI: 10.1002/prca.202400087] [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: 09/04/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
Determination of the changes in protein structure is crucial for a better understanding of their function and properties, which is highly important in identifying the causes of the disease, new drug development, and clinical applications. The Ramachandran plot, displaying the set of torsional angles, phi (Φ) and psi (Ψ), of the protein backbone, serves as a popular and convenient tool for secondary structure analysis and interpretation. However, identifying subtle changes in protein structure is often hindered in traditional Ramachandran plot, especially with the large amount of data generated by molecular dynamics (MD) simulations. In this paper, we proposed a useful and efficient tool, that is, differential Ramachandran plot (dRama), which enables to compare protein structures and extract the differences, providing a highly readable graphical representation. dRama is available at: https://github.com/MaksWolf44/dRama.
Collapse
Affiliation(s)
- Piotr Batys
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Krakow, Poland
| | - Leszek Krzemień
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Krakow, Poland
| | - Jakub Barbasz
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
19
|
Das RP, Behera SK, Sahoo B, Arakha M, Pradhan AK. Comparative analysis of backbone atom cross-correlation matrices and folding dynamics of amyloid fibril and its complexes with novel biosurfactants isolated from Bacillus strain: a binding free energy calculation (mM-PBSA) and MD simulation approach. J Biomol Struct Dyn 2024:1-16. [PMID: 39731748 DOI: 10.1080/07391102.2024.2446677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/25/2024] [Indexed: 12/30/2024]
Abstract
In the relentless pursuit of unraveling the intricate pathophysiology of Alzheimer's disease (AD), amyloid β (Aβ) proteins emerge as focal points due to their pivotal role in disease progression. The pathological hallmark of AD involves the aberrant aggregation of Aβ peptides into amyloid fibrils, precipitating a cascade of neurodegenerative events culminating in cognitive decline and neuronal loss. This study adopts a computational framework to investigate the potential therapeutic efficacy of novel biosurfactants (BS) in mitigating Aβ fibril formation. Initial analyses encompassing sequence alignment, structural elucidation, and functional characterization reveal distinctive attributes of the Aβ peptide and the identified BS candidates. Quantum chemical calculations, using the ORCA Program (v4.0) employed Density Functional Theory (DFT), specifically the Becke 3-parameter Lee-Yang-Parr (B3LYP) method, to investigate the electronic structure and energetics of novel isolates. Molecular docking through AutoDock Vina (version 1.1.2) employing advanced algorithms elucidates the binding affinities and interaction energies between Aβ fibrils and BS molecules. The observed binding energy of -7.0 kcal/mol for BG2A and -6.6 kcal/mol for BG2B, underscoring the robustness and stability of the formed complexes. The binding mechanism of docked complexes was predicted through molecular dynamics (MD) simulations using GROMACS 2021.3 and Charmm36 force field, capture complex dynamics over 100 nanoseconds. Analysis via RMSD, RMSF, Rg, PCA, and SASA offers insights into Aβ-BS complex stability and dynamics. These promising results highlight the potential of BG2A and BG2B as therapeutic candidates against AD. However, rigorous preclinical and clinical validation is crucial to ascertain their safety, efficacy, and translational relevance.
Collapse
Affiliation(s)
- Rohit Pritam Das
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India
| | - Santosh Kumar Behera
- Department of Biotechnology, National institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat, India
| | - Banishree Sahoo
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India
| | - Manoranjan Arakha
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India
| | - Arun Kumar Pradhan
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India
| |
Collapse
|
20
|
Liang Y, Zhang P, Liu M, Liu H, He B, Zhu Y, Wang J. Plant-based protein amyloid fibrils: Origins, formation, extraction, applications, and safety. Food Chem 2024; 469:142559. [PMID: 39732075 DOI: 10.1016/j.foodchem.2024.142559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/26/2024] [Accepted: 12/16/2024] [Indexed: 12/30/2024]
Abstract
Amyloid fibrils (AFs) are highly ordered nanostructures formed through the self-assembly of proteins under specific conditions. Due to their unique properties, AFs have garnered significant attention as biomaterials over the past decade. Nevertheless, the increasing reliance on animal proteins for AFs production raises sustainability concerns, highlighting the need for a transition to plant-based proteins as more environmentally friendly feedstocks. This review summarizes the conditions, mechanisms, and factors influencing the fibrillisation of over 20 plant-based protein amyloid fibrils (PAFs). The effectiveness of enzymatic extraction and membrane separation for isolating PAFs was also evaluated. Additionally, the review discusses the potential for enhancing PAFs' suitability through cross-linking with external agents. In the future, PAFs may be developed as advanced nanomaterials for a range of applications, including food hydrogels, cell-cultured meat scaffolds, and food detection sensors. However, thorough investigation of safety concerns and process improvements remain the primary challenges for the development of PAFs.
Collapse
Affiliation(s)
- Ying Liang
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China.
| | - Penghui Zhang
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China.
| | - Mei Liu
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China.
| | - Hao Liu
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Baoshan He
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Yingying Zhu
- College of Food and Bioengineering, Zhengzhou R&D Center for high-quality innovation of Green Food (Green Premium Agricultural Products), Zhengzhou University of Light Industry, Zhengzhou 450001, China
| | - Jinshui Wang
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China.
| |
Collapse
|
21
|
Choi SI, Jin Y, Choi Y, Seong BL. Beyond Misfolding: A New Paradigm for the Relationship Between Protein Folding and Aggregation. Int J Mol Sci 2024; 26:53. [PMID: 39795912 PMCID: PMC11720324 DOI: 10.3390/ijms26010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Aggregation is intricately linked to protein folding, necessitating a precise understanding of their relationship. Traditionally, aggregation has been viewed primarily as a sequential consequence of protein folding and misfolding. However, this conventional paradigm is inherently incomplete and can be deeply misleading. Remarkably, it fails to adequately explain how intrinsic and extrinsic factors, such as charges and cellular macromolecules, prevent intermolecular aggregation independently of intramolecular protein folding and structure. The pervasive inconsistencies between protein folding and aggregation call for a new framework. In all combined reactions of molecules, both intramolecular and intermolecular rate (or equilibrium) constants are mutually independent; accordingly, intrinsic and extrinsic factors independently affect both rate constants. This universal principle, when applied to protein folding and aggregation, indicates that they should be treated as two independent yet interconnected processes. Based on this principle, a new framework provides groundbreaking insights into misfolding, Anfinsen's thermodynamic hypothesis, molecular chaperones, intrinsic chaperone-like activities of cellular macromolecules, intermolecular repulsive force-driven aggregation inhibition, proteome solubility maintenance, and proteinopathies. Consequently, this paradigm shift not only refines our current understanding but also offers a more comprehensive view of how aggregation is coupled to protein folding in the complex cellular milieu.
Collapse
Affiliation(s)
- Seong Il Choi
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Vaccine Innovative Technology ALliance (VITAL)-Korea, Seoul 03722, Republic of Korea; (Y.J.); (Y.C.)
| | - Yoontae Jin
- Vaccine Innovative Technology ALliance (VITAL)-Korea, Seoul 03722, Republic of Korea; (Y.J.); (Y.C.)
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yura Choi
- Vaccine Innovative Technology ALliance (VITAL)-Korea, Seoul 03722, Republic of Korea; (Y.J.); (Y.C.)
- Department of Integrative Biotechnology, Yonsei University, Incheon 21983, Republic of Korea
| | - Baik L. Seong
- Vaccine Innovative Technology ALliance (VITAL)-Korea, Seoul 03722, Republic of Korea; (Y.J.); (Y.C.)
- Department of Microbiology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
22
|
Yong J, Villalta JE, Vu N, Kukurugya MA, Olsson N, López MP, Lazzari-Dean JR, Hake K, McAllister FE, Bennett BD, Jan CH. Impairment of lipid homeostasis causes lysosomal accumulation of endogenous protein aggregates through ESCRT disruption. eLife 2024; 12:RP86194. [PMID: 39713930 DOI: 10.7554/elife.86194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024] Open
Abstract
Protein aggregation increases during aging and is a pathological hallmark of many age-related diseases. Protein homeostasis (proteostasis) depends on a core network of factors directly influencing protein production, folding, trafficking, and degradation. Cellular proteostasis also depends on the overall composition of the proteome and numerous environmental variables. Modulating this cellular proteostasis state can influence the stability of multiple endogenous proteins, yet the factors contributing to this state remain incompletely characterized. Here, we performed genome-wide CRISPRi screens to elucidate the modulators of proteostasis state in mammalian cells, using a fluorescent dye to monitor endogenous protein aggregation. These screens identified known components of the proteostasis network and uncovered a novel link between protein and lipid homeostasis. Increasing lipid uptake and/or disrupting lipid metabolism promotes the accumulation of sphingomyelins and cholesterol esters and drives the formation of detergent-insoluble protein aggregates at the lysosome. Proteome profiling of lysosomes revealed ESCRT accumulation, suggesting disruption of ESCRT disassembly, lysosomal membrane repair, and microautophagy. Lipid dysregulation leads to lysosomal membrane permeabilization but does not otherwise impact fundamental aspects of lysosomal and proteasomal functions. Together, these results demonstrate that lipid dysregulation disrupts ESCRT function and impairs proteostasis.
Collapse
Affiliation(s)
- John Yong
- Calico Life Sciences LLC, South San Francisco, United States
| | | | - Ngoc Vu
- Calico Life Sciences LLC, South San Francisco, United States
| | | | - Niclas Olsson
- Calico Life Sciences LLC, South San Francisco, United States
| | | | | | - Kayley Hake
- Calico Life Sciences LLC, South San Francisco, United States
| | | | | | - Calvin H Jan
- Calico Life Sciences LLC, South San Francisco, United States
| |
Collapse
|
23
|
Wang W, Wu X, Zhang Q, Zhang T, Jiang L, Qu L, Lu F, Liu F. Tetrahydrofolic acid accelerates amyloid fibrillization, decreases cytotoxic oligomers and suppresses their toxicity. Int J Biol Macromol 2024; 290:139041. [PMID: 39708879 DOI: 10.1016/j.ijbiomac.2024.139041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/03/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Soluble cytotoxic oligomers produced during the fibrillation of both α-synuclein (αS) and amyloid-β protein (Aβ) are key pathogenic factors in Parkinson's disease (PD) and Alzheimer's disease (AD). Reducing toxic oligomers by regulating the aggregation process of αS and Aβ is an important strategy for the treatment of PD and AD. Herein, tetrahydrofolic acid (THF) is found to accelerate amyloid fibrillization, decreases cytotoxic oligomers and suppresses their toxicity. Thioflavin T and atomic force microscopy assays results showed that THF was able to accelerate the formation of dense fibrils from αS and Aβ in a dose-dependent manner. Strikingly, this was accompanied by a reduction in the abundance of toxic oligomers, and these results were confirmed by DB. Meanwhile, MTT and FDA/PI assays demonstrated that THF-induced accelerated fibril formation was accompanied by a reduction in αS- and Aβ-induced cytotoxicity. In addition, the lifespan of genetically modified αS and Aβ expressing C. elegans was extended by feeding THF, although plaque deposits of αS and Aβ increased. These findings suggest that THF enhances the conversion of αS and Aβ oligomers into less toxic fibrils and is a potential therapeutic agent for PD and AD.
Collapse
Affiliation(s)
- Wenqian Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Xinming Wu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Qingfu Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Tong Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Luying Jiang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Lili Qu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Fuping Lu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China
| | - Fufeng Liu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin 300457, PR China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, PR China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, PR China.
| |
Collapse
|
24
|
Misiu Naitė I, Mikalauskaitė K, Paulauskaitė M, Sniečkutė RT, Smirnovas V, Brukštus A, Žiaunys M, Žutautė I. Imidazo[2,1- b][1,3]thiazine Derivatives as Potential Modulators of Alpha-Synuclein Amyloid Aggregation. ACS Chem Neurosci 2024; 15:4418-4430. [PMID: 39603795 DOI: 10.1021/acschemneuro.4c00451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Insoluble amyloid fibrils accumulate in the intercellular spaces of organs and tissues, leading to various amyloidosis-related disorders in the human body. Specifically, Parkinson's disease is associated with the aggregation of alpha-synuclein. However, current treatments for Parkinson's primarily focus on managing motor symptoms and slowing disease progression. Efforts to prevent and halt the progression of these diseases involve the search for small molecular compounds. In this work, we synthesized imidazo[2,1-b][1,3]thiazines in an atom-economic way by cyclization of 2-alkynylthioimidazoles using 10% AuCl as the catalyst. We identified several compounds with specific functional groups capable of both inhibiting the aggregation of alpha-synuclein and redirecting the fibril formation pathway. The investigation into how these substances function revealed that imidazo[2,1-b][1,3]thiazine derivatives can influence alpha-synuclein aggregation in several ways. They not only inhibit the primary nucleation process and maintain a balance toward nonaggregated protein states but also stabilize smaller oligomeric species of alpha-synuclein and cause the formation of fibrils with unique structures and forms. These imidazo[2,1-b][1,3]thiazines could potentially be used in developing highly efficient, small molecular weight protein aggregation inhibitors.
Collapse
Affiliation(s)
- Indrė Misiu Naitė
- Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko st. 24, Vilnius LT-03225, Lithuania
| | - Kamilė Mikalauskaitė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, Vilnius LT-10257, Lithuania
| | - Martyna Paulauskaitė
- Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko st. 24, Vilnius LT-03225, Lithuania
| | - Ru Ta Sniečkutė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, Vilnius LT-10257, Lithuania
| | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, Vilnius LT-10257, Lithuania
| | - Algirdas Brukštus
- Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko st. 24, Vilnius LT-03225, Lithuania
| | - Mantas Žiaunys
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, Vilnius LT-10257, Lithuania
| | - Ieva Žutautė
- Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko st. 24, Vilnius LT-03225, Lithuania
| |
Collapse
|
25
|
Zhao Y, Brener O, Andrzejewska E, Wei J, Reiß C, Tietz O, Knowles TPJ, Aigbirhio FI. Detecting and Tracking β-Amyloid Oligomeric Forms and Dynamics In Vitro by a High-Sensitivity Fluorescent-Based Assay. ACS Chem Neurosci 2024; 15:4383-4389. [PMID: 39611283 DOI: 10.1021/acschemneuro.4c00312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
Aggregation of β-amyloid protein is a hallmark pathology of the neurodegenerative disorder Alzheimer's disease and proceeds from monomers to insoluble misfolded fibril forms via soluble and highly toxic oligomeric intermediates. Given the dual feature of being the most toxic form of the Aβ aggregate proteome and an early marker of pathogenesis, there is a need for sensitive methods that can be used to detect Aβ oligomers and investigate the dynamics of aggregation. Herein, we describe a method based on the application of an oligomer-sensitive fluorescent chemical probe pTP-TFE combined with the use of a QIAD (Quantitative determination of Interference with Aβ Aggregate Size Distribution) assay to correctly identify Aβ oligomers in high sensitivity. pTP-TFE was evaluated and compared to thioflavin T and pFTAA, the two most widely used amyloid fibril dyes, and shown to be the only probe capable of detecting significant differences across all oligomeric species of β-amyloid. Furthermore, by observing changes in pTP-TFE fluorescence emission over time, we could track the dynamics of oligomer populations and thereby obtain kinetic information on the Aβ42 dynamic aggregation model. Therefore, we have established a highly sensitive, readily available, and simple method for studying β-amyloid protein aggregation dynamics.
Collapse
Affiliation(s)
- Yanyan Zhao
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, U.K
| | - Oleksandr Brener
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf 40225, Germany
| | - Ewa Andrzejewska
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge,, Cambridge CB2 1EZ, U.K
| | - Jiapeng Wei
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge,, Cambridge CB2 1EZ, U.K
| | - CloudOuterMan Reiß
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Düsseldorf 40225, Germany
| | - Ole Tietz
- Dementia Research Centre, Department of Biomedical Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge,, Cambridge CB2 1EZ, U.K
| | - Franklin I Aigbirhio
- Molecular Imaging Chemistry Laboratory, Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0QQ, U.K
| |
Collapse
|
26
|
Shabnam, Bhat R. Flavones Suppress Aggregation and Amyloid Fibril Formation of Human Lysozyme under Macromolecular Crowding Conditions. Biochemistry 2024; 63:3194-3212. [PMID: 39385522 DOI: 10.1021/acs.biochem.4c00362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The crowded milieu of a biological cell significantly impacts protein aggregation and interactions. Understanding the effects of macromolecular crowding on the aggregation and fibrillation of amyloidogenic proteins is crucial for the treatment of many amyloid-related disorders. Most in vitro studies of protein amyloid formation and its inhibition by small molecules are conducted in dilute buffers, which do not mimic the complexity of the cellular environment. In this study, we used PEGs to simulate macromolecular crowding and examined the inhibitory effects of flavones DHF, baicalein, and luteolin on human lysozyme (HuL) aggregation at pH 2. Naturally occurring flavones have been effective inhibitors of amyloid formation in some proteins. Our findings indicate that while flavones inhibit HuL aggregation and fibrillation in dilute buffer solutions, complete inhibition is observed with a combination of flavones and PEGs, as shown by ThT fluorescence, light scattering, TEM, and AFM studies. The species formed in the presence of PEG 8000 and flavones were less hydrophobic, less toxic, and α-helix-rich compared to control samples, which were hydrophobic and β-sheet-rich, as demonstrated by ANS hydrophobicity, MTT assay, and CD spectroscopy. Fluorescence titration studies of flavones with HuL showed a significant increase in binding constant values under crowding conditions. These findings highlight the importance of macromolecular crowding in modulating protein aggregation and amyloid inhibition. Further studies using disease-causing mutants of HuL and other amyloidogenic proteins are needed to explore the role of macromolecular crowding in small-molecule-mediated modulation and inhibition of protein aggregation and amyloid formation.
Collapse
Affiliation(s)
- Shabnam
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 100067, India
| | - Rajiv Bhat
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 100067, India
| |
Collapse
|
27
|
Matveyenka M, Ali A, Mitchell CL, Sholukh M, Kurouski D. Elucidation of cytotoxicity of α-Synuclein fibrils on immune cells. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2024; 1873:141061. [PMID: 39694308 DOI: 10.1016/j.bbapap.2024.141061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/22/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Progressive aggregation of α-synuclein (α-Syn), a small cytosolic protein involved in cell vesicle trafficking, in the midbrain, hypothalamus, and thalamus is linked to Parkinson's disease (PD). Amyloid oligomers and fibrils formed as a result of such aggregation are highly toxic to neurons. However, it remains unclear whether amyloid-induced toxicity of neurons is the primary mechanism of the progressive neurodegeneration observed upon PD. In the current study, we investigated cytotoxicity exerted by α-Syn fibrils formed in the lipid-free environment, as well as in the presence of two phospholipids, on macrophages, dendritic cells, and microglia. We found that α-Syn fibrils are far more toxic to dendritic cells and microglia compared to neurons. We also observe low toxicity levels of such amyloids to macrophages. Real-time polymerase chain reaction (RT-PCR) results suggest that toxicity of amyloids aggregates is linked to the levels of autophagy in cells. These results suggest that a strong impairment of the immune system in the brain may be the first stop of neurodegenerative processes that are taking place upon the onset of PD.
Collapse
Affiliation(s)
- Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Abid Ali
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Charles L Mitchell
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Mikhail Sholukh
- Department of Biology, Belarussian State University, Minsk, 222000, Belarus
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, United States.
| |
Collapse
|
28
|
Fertan E, Hung C, Danial JSH, Lam JYL, Preman P, Albertini G, English EA, Böken D, Livesey FJ, De Strooper B, Patani R, Klenerman D. Clearance of beta-amyloid and tau aggregates is size dependent and altered by an inflammatory challenge. Brain Commun 2024; 7:fcae454. [PMID: 39749010 PMCID: PMC11694676 DOI: 10.1093/braincomms/fcae454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/12/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Extracellular beta-amyloid aggregation and inflammation are in a complex and not fully understood interplay during hyperphosphorylated tau aggregation and pathogenesis of Alzheimer's disease. Our group has previously shown that an immune challenge with tumour necrosis factor alpha can alter extracellular beta-sheet containing aggregates in human-induced pluripotent stem cell-derived cortical neurons carrying familial Alzheimer's disease-related presenilin 1 mutations. Here, using single-molecule detection and super-resolution imaging techniques, we quantified and characterized the intra- and extracellular beta-amyloid and AT8-positive tau aggregates. Our results indicate a pre-existing Alzheimer's disease-like pathology caused by the presenilin 1 mutation, with increased beta-amyloid aggregates in both the cell lysate and conditioned media compared to isogenic controls and also increased intracellular tau aggregates. The main effect of tumour necrosis factor alpha treatment on presenilin 1 neurons was the formation of larger intracellular beta-amyloid aggregates. In contrast, isogenic controls showed more significant changes with tumour necrosis factor alpha treatment with an increase in beta-amyloid aggregates in the media but not intracellularly and an increase in tau aggregates in both the media and cell lysate, suggesting a chronic inflammation-driven mechanism for the development of sporadic Alzheimer's disease. Remarkably, we also found significant morphological differences between intra- and extracellular beta-amyloid and tau aggregates in human-induced pluripotent stem cell-derived cortical neurons, suggesting these neurons can only clear aggregates when small, and that larger aggregates stay inside the neurons. While majority of the beta-amyloid aggregates were cleared into the media, a greater portion of the tau aggregates remained intracellular. This size-dependent aggregate clearance was also shown to be conserved in vivo, using soaked and homogenized mouse and human post-mortem Alzheimer's disease brain samples. As such, our results are proposing a previously unknown, size-dependent aggregate clearance mechanism, which can possibly explain the intracellular aggregation of tau and extracellular aggregation of beta-amyloid.
Collapse
Affiliation(s)
- Emre Fertan
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK
| | - Christy Hung
- The Francis Crick Institute, University College London, London NW1 1AT, UK
- Department of Neuroscience, City University of Hong Kong, Kowloon 999007, Hong Kong SAR
| | - John S H Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK
| | - Jeff Y L Lam
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK
| | - Pranav Preman
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 0N5 box 602, 3000 Leuven, Belgium
| | - Giulia Albertini
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 0N5 box 602, 3000 Leuven, Belgium
| | - Elizabeth A English
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK
| | - Dorothea Böken
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK
| | - Frederick J Livesey
- Zayed Centre for Research into Rare Disease in Children, University College London, Great Ormond Street Institute of Child Health, London WC1N 1DZ, UK
| | - Bart De Strooper
- VIB-KU Leuven Center for Brain & Disease Research, Herestraat 49, 0N5 box 602, 3000 Leuven, Belgium
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Rickie Patani
- The Francis Crick Institute, University College London, London NW1 1AT, UK
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0XY, UK
| |
Collapse
|
29
|
Minshull T, Byrd EJ, Olejnik M, Calabrese AN. Hydrogen-Deuterium Exchange Mass Spectrometry Reveals Mechanistic Insights into RNA Oligonucleotide-Mediated Inhibition of TDP-43 Aggregation. J Am Chem Soc 2024; 146:33626-33639. [PMID: 39610319 PMCID: PMC11638948 DOI: 10.1021/jacs.4c11229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Abstract
Deposits of aggregated TAR DNA-binding protein 43 (TDP-43) in the brain are associated with several neurodegenerative diseases. It is well established that binding of RNA/DNA to TDP-43 can prevent TDP-43 aggregation, but an understanding of the structure(s) and conformational dynamics of TDP-43, and TDP-43-RNA complexes, is lacking, including knowledge of how the solution environment modulates these properties. Here, we address this challenge using hydrogen-deuterium exchange-mass spectrometry. In the presence of RNA olignoucleotides, we observe protection from exchange in the RNA recognition motif (RRM) domains of TDP-43 and the linker region between the RRM domains, consistent with nucleic acid binding modulating interdomain interactions. Intriguingly, at elevated salt concentrations, the extent of protection from exchange is reduced in the RRM domains when bound to an RNA sequence derived from the 3' UTR of the TDP-43 mRNA (CLIP34NT) compared to when bound to a (UG)6 repeat sequence. Under these conditions, CLIP34NT is no longer able to prevent TDP-43 aggregation. This suggests that a salt-induced structural rearrangement occurs when bound to this RNA, which may play a role in facilitating aggregation. Additionally, upon RNA binding, we identify differences in exchange within the short α-helical region located in the C-terminal domain (CTD) of TDP-43. These allosterically altered regions may influence the ability of TDP-43 to aggregate and fine-tune its RNA binding repertoire. Combined, these data provide additional insights into the intricate interplay between TDP-43 aggregation and RNA binding, an understanding of which is crucial for unraveling the molecular mechanisms underlying TDP-43-associated neurodegeneration.
Collapse
Affiliation(s)
- Thomas
C. Minshull
- Astbury Centre for Structural
Molecular Biology, School of Molecular and Cellular Biology, Faculty
of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
| | - Emily J. Byrd
- Astbury Centre for Structural
Molecular Biology, School of Molecular and Cellular Biology, Faculty
of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
| | - Monika Olejnik
- Astbury Centre for Structural
Molecular Biology, School of Molecular and Cellular Biology, Faculty
of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
| | - Antonio N. Calabrese
- Astbury Centre for Structural
Molecular Biology, School of Molecular and Cellular Biology, Faculty
of Biological Sciences, University of Leeds, Leeds LS2 9JT, U.K.
| |
Collapse
|
30
|
Zhao D, Zhou Y, Xing F, Wang H, Zhou J. Porous organic cages as inhibitors of Aβ 42 peptide aggregation: a simulation study. Phys Chem Chem Phys 2024; 26:29696-29707. [PMID: 39501978 DOI: 10.1039/d4cp03549f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
The aggregation of Aβ monomers into oligomers with β-sheet structures is an important cause of Alzheimer's disease (AD), while the Aβ42 peptide is more toxic and prone to aggregate. It is of great significance to study the inhibition mechanism of Aβ42 monomer aggregation and find excellent inhibitors for the treatment of AD. Research in recent years has focused on small molecule compounds and nanoparticles, but they all have certain limitations. As a new type of porous material, a porous organic cage (POC) has potential application feasibility in the biomedical field due to its unique physicochemical properties. In this work, molecular dynamics simulations were used for the first time to explore the interaction and conformational transformation of the Aβ42 peptide in CC3 crystals with different morphologies (planar and spherical). The results show that the adsorption of the Aβ42 peptide on different CC3 crystals is mainly achieved through strong van der Waals forces. During the simulations, the Aβ42 peptide undergoes various degrees of structural changes. Compared to that in water, this binding induces more irregular structures, such as turns and 3-helices, and inhibits the production of β-sheets, while enhancing the overall backbone rigidity of the Aβ42 peptide. The transformation analysis of peptide conformation is further complemented by free energy landscape and cluster analysis. These findings provide a strong basis for CC3 crystals as novel inhibitors to inhibit the toxicity and aggregation of the Aβ42 peptide.
Collapse
Affiliation(s)
- Daohui Zhao
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, School of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, P. R. China.
| | - Yu Zhou
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, School of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, P. R. China.
| | - Fen Xing
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430060, P. R. China
| | - Hangxing Wang
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, School of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, P. R. China.
| | - Jian Zhou
- School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, P. R. China.
| |
Collapse
|
31
|
Ngah WZW, Ahmad HF, Ankasha SJ, Makpol S, Tooyama I. Dietary Strategies to Mitigate Alzheimer's Disease: Insights into Antioxidant Vitamin Intake and Supplementation with Microbiota-Gut-Brain Axis Cross-Talk. Antioxidants (Basel) 2024; 13:1504. [PMID: 39765832 PMCID: PMC11673287 DOI: 10.3390/antiox13121504] [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: 11/18/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 01/11/2025] Open
Abstract
Alzheimer's disease (AD), which is characterized by deterioration in cognitive function and neuronal death, is the most prevalent age-related progressive neurodegenerative disease. Clinical and experimental research has revealed that gut microbiota dysbiosis may be present in AD patients. The changed gut microbiota affects brain function and behavior through several mechanisms, including tau phosphorylation and increased amyloid deposits, neuroinflammation, metabolic abnormalities, and persistent oxidative stress. The lack of effective treatments to halt or reverse the progression of this disease has prompted a search for non-pharmaceutical tools. Modulation of the gut microbiota may be a promising strategy in this regard. This review aims to determine whether specific dietary interventions, particularly antioxidant vitamins, either obtained from the diet or as supplements, may support the formation of beneficial microbiota in order to prevent AD development by contributing to the systemic reduction of chronic inflammation or by acting locally in the gut. Understanding their roles would be beneficial as it may have the potential to be used as a future therapy option for AD patients.
Collapse
Affiliation(s)
- Wan Zurinah Wan Ngah
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| | - Hajar Fauzan Ahmad
- Department of Industrial Biotechnology, Faculty of Industrial Sciences and Technology, Universiti Malaysia Pahang Al-Sultan Abdullah, Gambang 26300, Pahang, Malaysia;
| | - Sheril June Ankasha
- Unisza Science and Medicine Foundation Centre, Universiti Sultan Zainal Abidin, Gong Badak Campus, Kuala Nerus 21300, Terengganu, Malaysia;
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Ikuo Tooyama
- Medical Innovation Research Center, Shiga University of Medical Science, Otsu 520-2192, Shiga, Japan;
| |
Collapse
|
32
|
Barclay AM, Milchberg MH, Warmuth OA, Tuttle MD, Dennis CJ, Schwieters CD, Rienstra CM. Automated fibril structure calculations in Xplor-NIH. Structure 2024:S0969-2126(24)00501-X. [PMID: 39662464 DOI: 10.1016/j.str.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/11/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
Amyloid fibrils are protein assemblies that are pathologically linked to neurodegenerative diseases. Fibril structures can aid development of highly specific ligands for diagnostic imaging and therapeutics. Solid-state NMR (SSNMR) is a viable approach to solving fibril structures; however, most SSNMR protocols require manual analysis of extensive spectral data, presenting a major bottleneck to determining structures. Standard automation; routines fall short for symmetric multimeric assemblies like amyloids due to high cross peak degeneracy and the need to account for multiple protein subunits. Here, we employ the probabilistic assignment for structure determination protocol in conjunction with strict; symmetry in Xplor-NIH structure determination software, demonstrating the methodology using data from a previous structure of an α-synuclein (Asyn) fibril implicated in Parkinson disease. The automated protocol generated a structure of comparable, if not superior, quality in a few days of computational time, reducing the manual effort required; to solve amyloid structures by SSNMR.
Collapse
Affiliation(s)
- Alexander M Barclay
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Moses H Milchberg
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Owen A Warmuth
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Marcus D Tuttle
- Department of Chemistry, Yale University, New Haven, CT 06511, USA
| | - Christopher J Dennis
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Charles D Schwieters
- Imaging Sciences Laboratory, Center for Information Technology, National Institutes of Health, Bethesda, MD 20817, USA
| | - Chad M Rienstra
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; National Magnetic Resonance Facility at Madison, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
33
|
Xu J, Sun H, Zhang Z, Guo Z, Le S, Chen H. Folding and Misfolding Dynamics of Irisin Protein Revealed by Single-Molecule Magnetic Tweezers. J Phys Chem Lett 2024; 15:11954-11960. [PMID: 39576132 DOI: 10.1021/acs.jpclett.4c02718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
Abstract
Irisin, a fibronectin III protein secreted by muscles during physical exercise, plays a significant role in the browning of white fat and cell adhesion, highlighting the importance of its conformational transitions. In this study, we investigated the folding and unfolding dynamics of a single irisin domain using a single-molecule manipulation technique known as magnetic tweezers. In addition to the native state, irisin can also fold transiently into a misfolded state. We determined the folding free energies of the native and misfolded states as well as their force-dependent folding and unfolding rates. The free energy of the misfolded state is higher than that of the unfolded state, and the misfolded state has a homogeneous force-dependent unfolding rate. The stable native state demonstrates heterogeneous unfolding rates that are within ∼1 order of magnitude. Via comparison with the well-studied 10th fibronectin III domain that has a partially folded intermediate state, our study demonstrates that proteins with similar structure can have distinct folding pathways.
Collapse
Affiliation(s)
- Jiashu Xu
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Department of Physics, Xiamen University, Xiamen 361005, China
| | - Hao Sun
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Department of Physics, Xiamen University, Xiamen 361005, China
- Center of Biomedical Physics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Zhuwei Zhang
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Department of Physics, Xiamen University, Xiamen 361005, China
| | - Zilong Guo
- Center of Biomedical Physics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Shimin Le
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Department of Physics, Xiamen University, Xiamen 361005, China
| | - Hu Chen
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Department of Physics, Xiamen University, Xiamen 361005, China
- Center of Biomedical Physics, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| |
Collapse
|
34
|
Dodero VI, Herrera MG. Oligomerization of 33-mer Gliadin Peptides: Supramolecular Assemblies in Celiac Disease. ChemMedChem 2024:e202400789. [PMID: 39635969 DOI: 10.1002/cmdc.202400789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/07/2024]
Abstract
The 33-mer gliadin peptide and its deamidated derivative, known as 33-mer DGP, are proteolytically resistant peptides central to the pathomechanism of celiac disease (CeD), the autoimmune presentation of gluten-related disorders (GRD). Both peptides can form spontaneous oligomers in the nanomolar concentration, leading to the formation of nanostructures. In other protein-related diseases, oligomers and aggregates are central in their pathomechanism; therefore, it was hypothesized that the oligomerization of proteolytical-resistant 33-mer gliadin peptides could be an underrecognized disease trigger. This review focuses on the current understanding of 33-mer peptides and their oligomers in vitro and cellular experiments. We intend to give the necessary details that incentivize the chemistry community to get involved in the effort to understand the self-assembly of gliadin peptides and the role of their supramolecular structures in CeD and the other GRD. More research is needed to design effective and safe chemical and/or nutritional interventions beyond the gluten-free diet.
Collapse
Affiliation(s)
- Verónica I Dodero
- Department of Chemistry, Bielefeld University, Universitätsstr. 25, 33615, Bielefeld, Germany
| | - María G Herrera
- Molecular Cell Biology, Faculty of Medicine, Ruhr University Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| |
Collapse
|
35
|
Beeg M, Rocutto B, Battocchio E, Dacomo L, Corbelli A, Fiordaliso F, Balducci C, Gobbi M. The Detection of Toxic Amyloid-β Fibril Fragments Through a Surface Plasmon Resonance Immunoassay. Int J Mol Sci 2024; 25:13020. [PMID: 39684731 DOI: 10.3390/ijms252313020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/28/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Amyloid-β1-42 (Aβ42) forms highly stable and insoluble fibrillar structures, representing the principal components of the amyloid plaques present in the brain of Alzheimer's disease (AD) patients. The involvement of Aβ42 in AD-associated neurodegeneration has also been demonstrated, in particular for smaller and soluble aggregates (oligomers). Based on these findings and on genetic evidence, Aβ42 aggregates are considered key players in the pathogenesis of AD and targets for novel therapies. Different approaches are currently used to detect the various aggregation states of Aβ peptide, including spectrophotometric methods, imaging techniques, and immunoassays, but all of these have specific limitations. To overcome them, we have recently exploited the peculiar properties of surface plasmon resonance (SPR) to develop an immunoassay capable of selectively detecting monomers and oligomers, discriminating them also from bigger fibrils in a mixture of different aggregated species, without any manipulation of the solution. In the present study, we extended these previous studies, showing that the SPR-based immunoassay makes it possible to unveil the fibril fragmentation induced mechanically, a result difficult to be conveniently and reliably assessed with other approaches. Moreover, we show that SPR-recognized fibril fragments are more toxic than the larger fibrillar structures, suggesting the relevance of the proposed SPR-based immunoassay.
Collapse
Affiliation(s)
- Marten Beeg
- Laboratory of Pharmacodynamics and Pharmacokinetics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via Mario Negri 2, 20156 Milan, Italy
| | - Beatrice Rocutto
- Laboratory of Pharmacodynamics and Pharmacokinetics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via Mario Negri 2, 20156 Milan, Italy
| | - Elisabetta Battocchio
- Laboratory of Pharmacodynamics and Pharmacokinetics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via Mario Negri 2, 20156 Milan, Italy
| | - Letizia Dacomo
- Laboratory of Biology of Neurodegenerative Disorders, Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via Mario Negri 2, 20156 Milan, Italy
| | - Alessandro Corbelli
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via Mario Negri 2, 20156 Milan, Italy
| | - Fabio Fiordaliso
- Laboratory of Molecular Biology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via Mario Negri 2, 20156 Milan, Italy
| | - Claudia Balducci
- Laboratory of Biology of Neurodegenerative Disorders, Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via Mario Negri 2, 20156 Milan, Italy
| | - Marco Gobbi
- Laboratory of Pharmacodynamics and Pharmacokinetics, Istituto di Ricerche Farmacologiche Mario Negri IRCCS Via Mario Negri 2, 20156 Milan, Italy
| |
Collapse
|
36
|
Chung HS. Characterizing heterogeneity in amyloid formation processes. Curr Opin Struct Biol 2024; 89:102951. [PMID: 39566372 PMCID: PMC11602362 DOI: 10.1016/j.sbi.2024.102951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/09/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
Protein aggregation is a complex process, consisting of a large number of pathways connecting monomers and mature amyloid fibrils. Recent advances in structure determination techniques, such as solid-state NMR and cryoEM, have allowed the determination of atomic resolution structures of fibril polymorphs, but most of the intermediate stages of the process including oligomer formation remain unknown. Proper characterization of the heterogeneity of the process is critical not only for physical and chemical understanding of the aggregation process but also for elucidation of the disease mechanisms and identification of therapeutic targets. This article reviews recent developments in the characterization of heterogeneity in amyloid formation processes.
Collapse
Affiliation(s)
- Hoi Sung Chung
- Laboratory of Chemical Physics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892-0520, USA.
| |
Collapse
|
37
|
Saade F, Barani C, Guyard M, Malatray M, Hugon-Vallet É, Schiele P. Amyloidosis and carpal tunnel syndrome: can we predict occurrence? HAND SURGERY & REHABILITATION 2024; 43:101789. [PMID: 39395761 DOI: 10.1016/j.hansur.2024.101789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
INTRODUCTION Carpal tunnel syndrome, especially when bilateral, may be associated with amyloid deposits, a precursor of systemic amyloidosis. Systemic amyloidosis affects various organs, cardiac involvement having the poorest prognosis. Diagnosis is often delayed. Current treatments are only effective in the early stages of the disease. The primary objective of our study was to evaluate the incidence of amyloidosis in patients with bilateral carpal tunnel syndrome; the secondary aim was to screen for cardiac forms. MATERIAL AND METHODS Between 2019 and 2023, we conducted a single-center prospective observational and diagnostic study to investigate the anatomical pathology of amyloid deposits in tenosynovial samples taken during open nerve release surgery on the median nerve. The tenosynovial biopsy was sent to a specific laboratory for analysis, and typing if positive. If amyloidosis was detected, the patient was referred to a specialist for a specific work-up. RESULTS We included 54 patients, with a mean age of 67 years (range, 51-89 years): 16 men and 38 women. Sixteen analyses were positive, 12 of which were transthyretin and 4 non-typed. DISCUSSION/CONCLUSION Bilateral carpal tunnel syndrome was predictive of amyloidosis. As synovial biopsy during surgery is simple and rapid, it should be implemented to identify amyloidosis at an early stage. This could change prognosis, by improving survival through screening and initiation of early specific treatment. LEVEL OF EVIDENCE IV.
Collapse
Affiliation(s)
- François Saade
- Service de Chirurgie Orthopédique - Hôpital Saint Joseph Saint Luc, 20 Quai Claude Bernard, 69007 Lyon, France.
| | - Camille Barani
- Service de Chirurgie Réparatrice et Esthétique - Hôpital Saint Joseph Saint Luc, 20 Quai Claude Bernard, 69007 Lyon, France
| | - Matthieu Guyard
- Service de Chirurgie Orthopédique - Hôpital Saint Joseph Saint Luc, 20 Quai Claude Bernard, 69007 Lyon, France
| | - Matthieu Malatray
- Service de Chirurgie Orthopédique - Hôpital Saint Joseph Saint Luc, 20 Quai Claude Bernard, 69007 Lyon, France
| | - Élisabeth Hugon-Vallet
- Service de Cardiologie - Hôpital Saint Joseph Saint Luc, 20 Quai Claude Bernard, 69007 Lyon, France
| | - Philippe Schiele
- Service de Chirurgie Orthopédique - Hôpital Saint Joseph Saint Luc, 20 Quai Claude Bernard, 69007 Lyon, France; Service d'Anatomopathologie - Hôpital Lyon Sud, 69495 Pierre Bénite, France
| |
Collapse
|
38
|
Iglesias V, Chilimoniuk J, Pintado-Grima C, Bárcenas O, Ventura S, Burdukiewicz M. Aggregating amyloid resources: A comprehensive review of databases on amyloid-like aggregation. Comput Struct Biotechnol J 2024; 23:4011-4018. [PMID: 39582896 PMCID: PMC11585477 DOI: 10.1016/j.csbj.2024.10.047] [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: 08/01/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 11/26/2024] Open
Abstract
Protein aggregation is responsible for several degenerative conditions in humans, and it is also a bottleneck in industrial protein production and storage of biotherapeutics. Bioinformatics tools have been developed to predict and redesign protein solubility more efficiently by understanding the underlying principles behind aggregation. As more experimental data become available, dedicated resources for storing, indexing, classifying and consolidating experimental results have emerged. These resources vary in focus, including aggregation-prone regions, 3D patches or protein stretches capable of forming amyloid fibrils. Some of these resources also consider the experimental conditions that cause protein aggregation and how they affect the process. This review article explores how protein aggregation databases have evolved and surveys state-of-the-art resources. We highlight their applications, complementarity and existing limitations. Moreover, we showcase the existing symbiosis between amyloid-related databases and predictive tools. To increase the usefulness of our review, we supplement it with a comprehensive list of present and past amyloid databases: https://biogenies.info/amyloid-database-list/.
Collapse
Affiliation(s)
- Valentín Iglesias
- Clinical Research Centre, Medical University of Białystok, Białystok, Poland
| | | | - Carlos Pintado-Grima
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Oriol Bárcenas
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Institute of Advanced Chemistry of Catalonia (IQAC), CSIC, Barcelona, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i de Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Hospital Universitari Parc Taulí, Institut d′Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Michał Burdukiewicz
- Clinical Research Centre, Medical University of Białystok, Białystok, Poland
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
39
|
Hekmat A, Kostova I, Saboury AA. Application of metallic nanoparticles-amyloid protein supramolecular materials in tissue engineering and drug delivery: Recent progress and perspectives. Colloids Surf B Biointerfaces 2024; 244:114185. [PMID: 39226848 DOI: 10.1016/j.colsurfb.2024.114185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
Supramolecular medicine refers to the formulation of therapeutic and diagnostic agents through supramolecular techniques, amid treating, diagnosing, and preventing disease. Recently, there has been growing interest in developing metal nanoparticles (MNPs)-amyloid hybrid materials, which have the potential to revolutionize medical applications. Furthermore, the development of MNPs-amyloid hydrogel/scaffold supramolecules represents a promising new direction in amyloid nanotechnology, with potential applications in tissue engineering and biomedicine. This review first provides a brief introduction to the formation process of protein amyloid aggregates and their unique nanostructures. Subsequently, we focused on recent investigations into the use of MNPs-amyloid hybrid materials in tissue engineering and biomedicine. We anticipate that MNPs-amyloid supramolecular materials will pave the way for new functional materials in medical science, particularly in the field of tissue engineering.
Collapse
Affiliation(s)
- Azadeh Hekmat
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Irena Kostova
- Faculty of Pharmacy, Medical University Sofia, Bulgaria
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
40
|
Kamalaldinezabadi SS, Watzlawik JO, Rosenberry TL, Paravastu AK, Stagg SM. Aggregation dynamics of a 150 kDa Aβ42 oligomer: Insights from cryo electron microscopy and multimodal analysis. Comput Struct Biotechnol J 2024; 23:4205-4213. [PMID: 39650331 PMCID: PMC11621449 DOI: 10.1016/j.csbj.2024.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 12/11/2024] Open
Abstract
Protein misfolding is a widespread phenomenon that can result in the formation of protein aggregates, which are markers of various disease states, including Alzheimer's disease (AD). In AD, amyloid beta (Aβ) peptides are key players in the disease's progression, particularly the 40- and 42- residue variants, Aβ40 and Aβ42. These peptides aggregate to form amyloid plaques and contribute to neuronal toxicity. Recent research has shifted attention from solely Aβ fibrils to also include Aβ protofibrils and oligomers as potentially critical pathogenic agents. Particularly, oligomers demonstrate more significant toxicity compared to other Aβ specie. Hence, there is an increased interest in studying the correlation between toxicity and their structure and aggregation pathway. The present study investigates the aggregation of a 150 kDa Aβ42 oligomer that does not lead to fibril formation. Using negative stain transmission electron microscopy (TEM), size exclusion chromatography (SEC), dynamic light scattering (DLS), and cryo-electron microscopy (cryo-EM), we demonstrate that 150 kDa Aβ42 oligomers form higher-order string-like assemblies over time. These strings are unique from the classical Aβ fibrils. The significance of our work lies in elucidating molecular behavior of a novel non-fibrillar form of Aβ42 aggregate.
Collapse
Affiliation(s)
| | - Jens O. Watzlawik
- The Departments on Neuroscience and Pharmacology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Terrone L. Rosenberry
- The Departments on Neuroscience and Pharmacology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Anant K. Paravastu
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Scott M. Stagg
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA
- Department of Biological Sciences, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
41
|
Otawa M, Itoh SG, Okumura H. Nonequilibrium Molecular Dynamics Method to Generate Poiseuille-Like Flow between Lipid Bilayers. J Chem Theory Comput 2024; 20:10199-10208. [PMID: 39526585 DOI: 10.1021/acs.jctc.4c00750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
There are various flows inside and outside cells in vivo. Nonequilibrium molecular dynamics (NEMD) simulation is a useful tool for understanding the effects of these flows on the dynamics of biomolecules. We propose an NEMD method to generate a Poiseuille-like flow between lipid bilayers. We extended the conventional equilibrium MD method to produce a flow by adding constant external force terms to the water molecules. Using the Lagrange multiplier method, the center of mass of the lipid bilayer is constrained so that the flow does not sweep away the lipid bilayer, but the individual lipid molecules fluctuate. The temperature of the system is controlled properly in the solution and membrane by using the Nosé-Hoover thermostat. We found that the flow velocity increases linearly as the applied external force term increases. It is possible to estimate the appropriate value of acceleration to generate a flow with an arbitrary velocity using this proportional relation once a single short MD simulation is performed. We also found that the flow between two lipid bilayers is slower than the analytical solution of the Navier-Stokes equations between rigid parallel plates due to the interactions between water molecules and the membrane. This method can be applied not only to a flow on lipid membranes but also to a flow on soft surfaces generally.
Collapse
Affiliation(s)
- Masaki Otawa
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
- Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Satoru G Itoh
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
- Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Hisashi Okumura
- Department of Structural Molecular Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Aichi 444-8787, Japan
- Institute for Molecular Science, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
42
|
Kamat S, Mayor T. Lost in translation: Threonine dictates aggregation of CAT tails. Mol Cell 2024; 84:4259-4261. [PMID: 39577399 DOI: 10.1016/j.molcel.2024.10.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/24/2024]
Abstract
In this issue of Molecular Cell, Chang et al.1 elaborate on the mechanisms by which CAT tail aggregation is mediated through threonine residues and how it contributes to the perturbation of proteostasis.
Collapse
Affiliation(s)
- Shriya Kamat
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; The Edwin S.H. Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC, Canada
| | - Thibault Mayor
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; The Edwin S.H. Leong Centre for Healthy Aging, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
43
|
Yagi-Utsumi M, Kanaoka Y, Miyajima S, Itoh SG, Yanagisawa K, Okumura H, Uchihashi T, Kato K. Single-Molecule Kinetic Observation of Antibody Interactions with Growing Amyloid β Fibrils. J Am Chem Soc 2024; 146:31518-31528. [PMID: 39445702 PMCID: PMC11583206 DOI: 10.1021/jacs.4c08841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Understanding the dynamic assembly process of amyloid β (Aβ) during fibril formation is essential for developing effective therapeutic strategies against Alzheimer's disease. Here, we employed high-speed atomic force microscopy to observe the growth of Aβ fibrils at the single-molecule level, focusing specifically on their interaction with anti-Aβ antibodies. Our findings show that fibril growth consists of intermittent periods of elongation and pausing, which are dictated by the alternating addition of Aβ monomers to protofilaments. We highlight the distinctive interaction of antibody 4396C, which specifically binds to the fibril ends in the paused state, suggesting a unique mechanism to hinder fibril elongation. Through real-time visualization of fibril growth and antibody interactions combined with molecular simulation, this study provides a refined understanding of Aβ assembly during fibril formation and suggests novel strategies for Alzheimer's therapy aimed at inhibiting the fibril elongation.
Collapse
Affiliation(s)
- Maho Yagi-Utsumi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi 465-8603, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
- Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Yui Kanaoka
- Department of Physics, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Shogo Miyajima
- Department of Physics, Nagoya University, Nagoya, Aichi 464-8602, Japan
| | - Satoru G Itoh
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
- Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Katsuhiko Yanagisawa
- Research and Development Center for Precision Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8550, Japan
- Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan
| | - Hisashi Okumura
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
- Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Takayuki Uchihashi
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
- Department of Physics, Nagoya University, Nagoya, Aichi 464-8602, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Aichi 464-0814, Japan
| | - Koichi Kato
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi 465-8603, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
- Institute for Molecular Science (IMS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
44
|
Jiang LL, Zhang XL, Hu HY. Co-Aggregation of TDP-43 with Other Pathogenic Proteins and Their Co-Pathologies in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:12380. [PMID: 39596445 PMCID: PMC11594478 DOI: 10.3390/ijms252212380] [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: 10/24/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Pathological aggregation of a specific protein into insoluble aggregates is a common hallmark of various neurodegenerative diseases (NDDs). In the earlier literature, each NDD is characterized by the aggregation of one or two pathogenic proteins, which can serve as disease-specific biomarkers. The aggregation of these specific proteins is thought to be a major cause of or deleterious result in most NDDs. However, accumulating evidence shows that a pathogenic protein can interact and co-aggregate with other pathogenic proteins in different NDDs, thereby contributing to disease onset and progression synergistically. During the past years, more than one type of NDD has been found to co-exist in some individuals, which may increase the complexity and pathogenicity of these diseases. This article reviews and discusses the biochemical characteristics and molecular mechanisms underlying the co-aggregation and co-pathologies associated with TDP-43 pathology. The TDP-43 aggregates, as a hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), can often be detected in other NDDs, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and spinocerebellar ataxia type 2 (SCA2). In many cases, TDP-43 is shown to interact and co-aggregate with multiple pathogenic proteins in vitro and in vivo. Furthermore, the co-occurrence and co-aggregation of TDP-43 with other pathogenic proteins have important consequences that may aggravate the diseases. Thus, the current viewpoint that the co-aggregation of TDP-43 with other pathogenic proteins in NDDs and their relevance to disease progression may gain insights into the patho-mechanisms and therapeutic potential of various NDDs.
Collapse
Affiliation(s)
- Lei-Lei Jiang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
| | - Xiang-Le Zhang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong-Yu Hu
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China; (L.-L.J.); (X.-L.Z.)
| |
Collapse
|
45
|
Tammara V, Doke AA, Jha SK, Das A. Deciphering the Monomeric and Dimeric Conformational Landscapes of the Full-Length TDP-43 and the Impact of the C-Terminal Domain. ACS Chem Neurosci 2024. [PMID: 39548975 DOI: 10.1021/acschemneuro.4c00557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2024] Open
Abstract
The aberrant aggregation of TAR DNA-binding protein 43 kDa (TDP-43) in cells leads to the pathogenesis of multiple fatal neurodegenerative diseases. Decoding the proposed initial transition between its functional dimeric and aggregation-prone monomeric states can potentially design a viable therapeutic strategy, which is presently limited by the lack of structural detail of the full-length TDP-43. To achieve a complete understanding of such a delicate phase space, we employed a multiscale simulation approach that unearths numerous crucial features, broadly summarized in two categories: (1) state-independent features that involve inherent chain collapsibility, rugged polymorphic landscape dictated by the terminal domains, high β-sheet propensity, structural integrity preserved by backbone-based intrachain hydrogen bonds and electrostatic forces, the prominence of the C-terminal domain in the intrachain cross-domain interfaces, and equal participation of hydrophobic and hydrophilic (charged and polar) residues in cross-domain interfaces; and (2) dimerization-modulated characteristics that encompass slower collapsing dynamics, restricted polymorphic landscape, the dominance of side chains in interchain hydrogen bonds, the appearance of the N-terminal domain in the dimer interface, and the prominence of hydrophilic (specifically polar) residues in interchain homo- and cross-domain interfaces. In our work, the ill-known C-terminal domain appears as the most crucial structure-dictating domain, which preferably populates a compact conformation with a high β-sheet propensity in its isolated state stabilized by intrabackbone hydrogen bonds, and these signatures are comparatively faded in its integrated form. Validation of our simulated observables by a complementary spectroscopic approach on multiple counts ensures the robustness of the computationally predicted features of the TDP-43 aggregation landscape.
Collapse
Affiliation(s)
- Vaishnavi Tammara
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Abhilasha A Doke
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh Kumar Jha
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Atanu Das
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
46
|
Kalmouni M, Oh Y, Alata W, Magzoub M. Designed Cell-Penetrating Peptide Constructs for Inhibition of Pathogenic Protein Self-Assembly. Pharmaceutics 2024; 16:1443. [PMID: 39598566 PMCID: PMC11597747 DOI: 10.3390/pharmaceutics16111443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
Peptides possess a number of pharmacologically desirable properties, including greater chemical diversity than other biomolecule classes and the ability to selectively bind to specific targets with high potency, as well as biocompatibility, biodegradability, and ease and low cost of production. Consequently, there has been considerable interest in developing peptide-based therapeutics, including amyloid inhibitors. However, a major hindrance to the successful therapeutic application of peptides is their poor delivery to target tissues, cells or subcellular organelles. To overcome these issues, recent efforts have focused on engineering cell-penetrating peptide (CPP) antagonists of amyloidogenesis, which combine the attractive intrinsic properties of peptides with potent therapeutic effects (i.e., inhibition of amyloid formation and the associated cytotoxicity) and highly efficient delivery (to target tissue, cells, and organelles). This review highlights some promising CPP constructs designed to target amyloid aggregation associated with a diverse range of disorders, including Alzheimer's disease, transmissible spongiform encephalopathies (or prion diseases), Parkinson's disease, and cancer.
Collapse
Affiliation(s)
| | | | | | - Mazin Magzoub
- Biology Program, Division of Science, New York University Abu Dhabi, Saadiyat Island Campus, Abu Dhabi P.O. Box 129188, United Arab Emirates; (Y.O.)
| |
Collapse
|
47
|
Doke AA, Jha SK. Identification of a Hidden, Highly Aggregation-Prone Intermediate of Full-Length TDP-43 That Triggers its Misfolding and Amyloid Aggregation. Biochemistry 2024. [PMID: 39530145 DOI: 10.1021/acs.biochem.4c00389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
In cells, TDP-43 is a crucial protein that can form harmful amyloid aggregates linked to fatal and incurable human neurodegenerative disorders. Normally, TDP-43 exists in a smaller soluble native state that prevents aggregation. However, aging and stress can destabilize this native state, leading to the formation of disease-causing amyloid aggregates via the formation of partially unfolded, high-energy intermediates with a greater tendency to aggregate. These intermediates are crucial in the early stages of amyloid formation and are challenging to study due to their low stability. Understanding the structure of these early aggregation-prone states of TDP-43 is essential for designing effective treatments for TDP-43 proteinopathies. Targeting these initial intermediates could be more effective than focusing on fully formed amyloid aggregates. By disrupting the aggregation process at this early stage, we may be able to prevent the progression of diseases related to TDP-43 aggregation. Hence, we decided to uncover the hidden, high-energy intermediates in equilibrium with the native states of TDP-43 by modulating the thermodynamic stability of the soluble native dimer (N form) and monomeric molten globular state (MG form) of full-length TDP-43. The thermodynamic modulation performed in the current study successfully revealed the highly aggregation-prone intermediate of full-length TDP-43, i.e., PUF. Moreover, we observed that along with high aggregation propensity, the aggregation kinetics and mechanisms of PUF differ from previously identified intermediates of full-length TDP-43 (the MG and I forms). The information regarding the initial aggregation-prone state of full-length TDP-43 could lead to therapies for amyloid diseases by halting early protein aggregation.
Collapse
Affiliation(s)
- Abhilasha A Doke
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh Kumar Jha
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
48
|
Zamarra GB, Sandu M, Caione N, Di Pasquale G, Di Berardino A, Di Ludovico A, La Bella S, Chiarelli F, Cattivera V, Colella J, Di Donato G. Amyloidosis in Childhood: A Review of Clinical Features and Comparison with Adult Forms. J Clin Med 2024; 13:6682. [PMID: 39597824 PMCID: PMC11594867 DOI: 10.3390/jcm13226682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Amyloidosis is a rare multisystem disorder characterized by extracellular accumulation of insoluble fibrils in various organs and tissues. The most common subtype in the pediatric population is systemic reactive amyloidosis, typically developing secondary to chronic inflammatory conditions and resulting in deposition of serum amyloid A protein in association with apolipoprotein HDL3. Clinical presentation is highly variable and is mostly influenced by specific organs involved, precursor protein type, and extent of amyloid deposition, often closely reflecting clinical features of the underlying disease. The most critical determinants of prognosis are cardiac and renal involvement. Diagnosis of amyloidosis is confirmed by tissue biopsy, which remains the gold standard, followed by precise amyloid fibril typing. The primary therapeutic approach is directed towards controlling underlying disease and reducing serum levels of precursor proteins to prevent further amyloid deposition. This study aims to highlight the main clinical characteristics of amyloidosis with onset in childhood, emphasizing the key differences compared to adult form.
Collapse
Affiliation(s)
- Giovanni Battista Zamarra
- Department of Pediatrics, L’Aquila University—UNIVAQ, 67100 L’Aquila, Italy; (G.B.Z.); (M.S.); (N.C.); (G.D.P.); (A.D.B.); (V.C.); (J.C.)
| | - Marina Sandu
- Department of Pediatrics, L’Aquila University—UNIVAQ, 67100 L’Aquila, Italy; (G.B.Z.); (M.S.); (N.C.); (G.D.P.); (A.D.B.); (V.C.); (J.C.)
| | - Nicholas Caione
- Department of Pediatrics, L’Aquila University—UNIVAQ, 67100 L’Aquila, Italy; (G.B.Z.); (M.S.); (N.C.); (G.D.P.); (A.D.B.); (V.C.); (J.C.)
| | - Gabriele Di Pasquale
- Department of Pediatrics, L’Aquila University—UNIVAQ, 67100 L’Aquila, Italy; (G.B.Z.); (M.S.); (N.C.); (G.D.P.); (A.D.B.); (V.C.); (J.C.)
| | - Alessio Di Berardino
- Department of Pediatrics, L’Aquila University—UNIVAQ, 67100 L’Aquila, Italy; (G.B.Z.); (M.S.); (N.C.); (G.D.P.); (A.D.B.); (V.C.); (J.C.)
| | - Armando Di Ludovico
- Department of Pediatrics, “G. D’Annunzio” University, 66100 Chieti, Italy; (A.D.L.); (S.L.B.); (F.C.)
| | - Saverio La Bella
- Department of Pediatrics, “G. D’Annunzio” University, 66100 Chieti, Italy; (A.D.L.); (S.L.B.); (F.C.)
| | - Francesco Chiarelli
- Department of Pediatrics, “G. D’Annunzio” University, 66100 Chieti, Italy; (A.D.L.); (S.L.B.); (F.C.)
| | - Valentina Cattivera
- Department of Pediatrics, L’Aquila University—UNIVAQ, 67100 L’Aquila, Italy; (G.B.Z.); (M.S.); (N.C.); (G.D.P.); (A.D.B.); (V.C.); (J.C.)
| | - Jacopo Colella
- Department of Pediatrics, L’Aquila University—UNIVAQ, 67100 L’Aquila, Italy; (G.B.Z.); (M.S.); (N.C.); (G.D.P.); (A.D.B.); (V.C.); (J.C.)
| | - Giulio Di Donato
- Department of Pediatrics, L’Aquila University—UNIVAQ, 67100 L’Aquila, Italy; (G.B.Z.); (M.S.); (N.C.); (G.D.P.); (A.D.B.); (V.C.); (J.C.)
| |
Collapse
|
49
|
Seychell RM, El Saghir A, Vassallo N. Modulation of Biological Membranes Using Small-Molecule Compounds to Counter Toxicity Caused by Amyloidogenic Proteins. MEMBRANES 2024; 14:231. [PMID: 39590617 PMCID: PMC11596372 DOI: 10.3390/membranes14110231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
The transition of peptides or proteins along a misfolding continuum from soluble functional states to pathological aggregates, to ultimately deposit as amyloid fibrils, is a process that underlies an expanding group of human diseases-collectively known as protein-misfolding disorders (PMDs). These include common and debilitating conditions, such as Alzheimer's disease, Parkinson's disease, and type-2 diabetes. Compelling evidence has emerged that the complex interplay between the misfolded proteins and biological membranes is a key determinant of the pathogenic mechanisms by which harmful amyloid entities are formed and exert their cytotoxicity. Most efforts thus far to develop disease-modifying treatments for PMDs have largely focused on anti-aggregation strategies: to neutralise, or prevent the formation of, toxic amyloid species. Herein, we review the critical role of the phospholipid membrane in mediating and enabling amyloid pathogenicity. We consequently propose that the development of small molecules, which have the potential to uniquely modify the physicochemical properties of the membrane and make it more resilient against damage by misfolded proteins, could provide a novel therapeutic approach in PMDs. By way of an example, natural compounds shown to intercalate into lipid bilayers and inhibit amyloid-lipid interactions, such as the aminosterols, squalamine and trodusquamine, cholesterol, ubiquinone, and select polyphenols, are discussed. Such a strategy would provide a novel approach to counter a wide range of toxic biomolecules implicit in numerous human amyloid pathologies.
Collapse
Affiliation(s)
- Raina Marie Seychell
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Adam El Saghir
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
| | - Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, MSD 2080 Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, MSD 2080 Msida, Malta
| |
Collapse
|
50
|
Tang Z, Fang Z, Wu X, Liu J, Tian L, Li X, Diao J, Ji B, Li D. Folding of N-terminally acetylated α-synuclein upon interaction with lipid membranes. Biophys J 2024; 123:3698-3720. [PMID: 39306670 PMCID: PMC11560312 DOI: 10.1016/j.bpj.2024.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
α-Synuclein (α-syn) is an abundant presynaptic neuronal protein whose aggregation is strongly associated with Parkinson's disease. It has been proposed that lipid membranes significantly affect α-syn's aggregation process. Extensive studies have been conducted to understand the interactions between α-syn and lipid membranes and have demonstrated that the N-terminus plays a critical role. However, the dynamics of the interactions and the conformational transitions of the N-terminus of α-syn at the atomistic scale details are still highly desired. In this study, we performed extensive enhanced sampling molecular dynamics simulations to quantify the folding and interactions of wild-type and N-terminally acetylated α-syn when interacting with lipid structures. We found that N-terminal acetylation significantly increases the helicity of the first few residues in solution or when interacting with lipid membranes. The observations in simulations showed that the binding of α-syn with lipid membranes mainly follows the induced-fit model, where the disordered α-syn binds with the lipid membrane through the electrostatic interactions and hydrophobic contacts with the packing defects; after stable insertion, N-terminal acetylation promotes the helical folding of the N-terminus to enhance the anchoring, thus increasing the binding affinity. We have shown the critical role of the first N-terminal residue methionine for recognition and anchoring to the negatively charged membrane. Although N-terminal acetylation neutralizes the positive charge of Met1 that may affect the electrostatic interactions of α-syn with membranes, the increase in helicity of the N-terminus should compensate for the binding affinity. This study provides detailed insight into the folding dynamics of α-syn's N-terminus with or without acetylation in solution and upon interaction with lipids, which clarifies how the N-terminal acetylation regulates the affinity of α-syn binding to lipid membranes. It also shows how packing defects and electrostatic effects coregulate the N-terminus of α-syn folding and its interaction with membranes.
Collapse
Affiliation(s)
- Zihan Tang
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Zhou Fang
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Xuwei Wu
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Jie Liu
- MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Liangfei Tian
- MOE Key Laboratory of Biomedical Engineering, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Xuejin Li
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Baohua Ji
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health) and Wenzhou Institute of University of Chinese Academy of Science, Wenzhou, China
| | - Dechang Li
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Institute of Biomechanics and Applications, Department of Engineering Mechanics, Zhejiang University, Hangzhou, China.
| |
Collapse
|