51
|
Gatch AJ, Ding F. Cross-Interaction with Amyloid-β Drives Pathogenic Structural Transformation within the Amyloidogenic Core Region of TDP-43. ACS Chem Neurosci 2025; 16:1565-1581. [PMID: 40167418 PMCID: PMC12003063 DOI: 10.1021/acschemneuro.5c00084] [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] [Indexed: 04/02/2025] Open
Abstract
Alzheimer's disease (AD) is the world's most prevalent neurodegenerative disorder, characterized neuropathologically by senile plaques and neurofibrillary tangles formed by amyloid-β (Aβ) and tau, respectively. Notably, a subset of AD patients also exhibits pathological aggregates composed of TAR DNA-Binding Protein 43 (TDP-43). Clinically, the presence of TDP-43 copathology in AD correlates with more severe cognitive decline and faster disease progression. While previous studies have shown that TDP-43 can exacerbate Aβ toxicity and modulate its assembly dynamics by delaying fibrillization and promoting oligomer formation, the impact of the Aβ interaction on the structural dynamics and aggregation of TDP-43 remains unclear. Here, we employed all-atom discrete molecular dynamics simulations to study the direct interaction between Aβ42, the more amyloidogenic isoform of Aβ, and the amyloidogenic core region (ACR) of TDP-43, which spans residues 311-360 and is critical for TDP-43 aggregation. We found that monomeric Aβ42 could strongly bind to the ACR, establishing sustained contact through intermolecular hydrogen bonding. In contrast, simulation of ACR dimerization revealed a transient helix-helix interaction, experimentally known to drive the phase separation behavior of TDP-43. The binding of the ACR to an Aβ42 fibril seed resulted in significant structural transformation, with the complete unfolding of the helical region being observed. Furthermore, interaction with the Aβ42 fibril seed catalyzed the formation of a parallel, in-register intermolecular β-sheet between two ACR monomers. Collectively, our computational study provides important theoretical insights into TDP-43 pathology in AD, demonstrating that Aβ42, especially in its fibrillar form, may catalyze the pathogenic structural transformation within the TDP-43 ACR that initiates its aberrant aggregation.
Collapse
Affiliation(s)
- Adam J. Gatch
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, United States
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| |
Collapse
|
52
|
Cathoud G, Hashemi M, Lyubchenko Y, Simões P. Uncovering Amyloid-β Interactions: Gray versus White Matter. ACS Chem Neurosci 2025; 16:1433-1441. [PMID: 40143654 DOI: 10.1021/acschemneuro.4c00439] [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: 03/28/2025] Open
Abstract
Alzheimer's disease is characterized by the accumulation of amyloid plaques in the brain. Recent studies suggest that amyloid-β (Aβ) peptides interact with cell membranes, potentially catalyzing plaque formation. However, the effect of varying cell membrane compositions on this catalytic process requires further investigation. Using molecular dynamics simulations, we demonstrate that a model gray matter membrane significantly influences the secondary structure of β-amyloid peptides. Notably, residues Asp1 and Glu22 play crucial roles in the membrane interaction. Glutamic acid at position 22, located in the middle of the peptide chain, appears to promote the formation of β-hairpin conformations, which are critical for aggregation. Additionally, our simulations reveal that the model white matter membrane allows a spontaneous insertion of segments of the peptide into the membrane, suggesting that membrane interaction not only alters the peptide structure but may also compromise membrane integrity. Our results show that the different membrane compositions in the brain may play different roles when interacting with β-amyloid peptides.
Collapse
Affiliation(s)
- Gabriel Cathoud
- CERES, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Pólo II, 3030-790 Coimbra, Portugal
| | - Mohtadin Hashemi
- Department of Physics, Auburn University, Auburn, Alabama 36849-5318, United States
| | - Yuri Lyubchenko
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198-6025, United States
| | - Pedro Simões
- CERES, Department of Chemical Engineering, University of Coimbra, Rua Sílvio Lima-Pólo II, 3030-790 Coimbra, Portugal
| |
Collapse
|
53
|
Brzezinski M, Argudo PG, Scheidt T, Yu M, Hosseini E, Kaltbeitzel A, Lemke EA, Michels JJ, Parekh SH. Protein-Specific Crowding Accelerates Aging in Protein Condensates. Biomacromolecules 2025; 26:2060-2075. [PMID: 39648588 DOI: 10.1021/acs.biomac.4c00609] [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: 12/10/2024]
Abstract
Macromolecular crowding agents, such as poly(ethylene glycol) (PEG), are often used to mimic cellular cytoplasm in protein assembly studies. Despite the perception that crowding agents have an inert nature, we demonstrate and quantitatively explore the diverse effects of PEG on the phase separation and maturation of protein condensates. We use two model proteins, the FG domain of Nup98 and bovine serum albumin (BSA), which represent an intrinsically disordered protein and a protein with a well-established secondary structure, respectively. PEG expedites the maturation of Nup98, enhancing denser protein packing and fortifying interactions, which hasten beta-sheet formation and subsequent droplet gelation. In contrast to BSA, PEG enhances droplet stability and limits the available solvent for protein solubilization, inducing only minimal changes in the secondary structure, pointing toward a significantly different role of the crowding agent. Strikingly, we detect almost no presence of PEG in Nup droplets, whereas PEG is moderately detectable within BSA droplets. Our findings demonstrate a nuanced interplay between crowding agents and proteins; PEG can accelerate protein maturation in liquid-liquid phase separation systems, but its partitioning and effect on protein structure in droplets is protein specific. This suggests that crowding phenomena are specific to each protein-crowding agent pair.
Collapse
Affiliation(s)
- Mateusz Brzezinski
- Department of Biomedical Engineering University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, Texas 78712, United States
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Pablo G Argudo
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Tom Scheidt
- Biocenter, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 17, Mainz 55128, Germany
- Institute of Molecular Biology GmbH, Ackermannweg 4, Mainz 55128, Germany
| | - Miao Yu
- Biocenter, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 17, Mainz 55128, Germany
- Institute of Molecular Biology GmbH, Ackermannweg 4, Mainz 55128, Germany
| | - Elnaz Hosseini
- Department of Biomedical Engineering University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, Texas 78712, United States
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Anke Kaltbeitzel
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Edward A Lemke
- Biocenter, Johannes Gutenberg University Mainz, Hanns-Dieter-Hüsch-Weg 17, Mainz 55128, Germany
- Institute of Molecular Biology GmbH, Ackermannweg 4, Mainz 55128, Germany
| | - Jasper J Michels
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| | - Sapun H Parekh
- Department of Biomedical Engineering University of Texas at Austin, 107 W. Dean Keeton Rd., Austin, Texas 78712, United States
- Max Planck Institute for Polymer Research Ackermannweg 10, Mainz 55128, Germany
| |
Collapse
|
54
|
Cavallaro F, Conti Nibali S, Cubisino SAM, Caruso P, Zimbone S, Infantino IR, Reina S, De Pinto V, Messina A, Giuffrida ML, Magrì A. VDAC1-Targeted NHK1 Peptide Recovers Mitochondrial Dysfunction Counteracting Amyloid-β Oligomers Toxicity in Alzheimer's Disease. Aging Cell 2025:e70069. [PMID: 40223243 DOI: 10.1111/acel.70069] [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: 10/19/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Mitochondrial dysfunction has been implicated in a broad range of age-related pathologies and has been proposed as a causative factor in Alzheimer's disease (AD). Analysis of post-mortem brains from AD patients showed increased levels of Voltage-dependent anion-selective channel 1 (VDAC1) in the dystrophic neurites surrounding amyloid-β (Aβ) deposits, suggesting a direct association between VDAC1 and mitochondrial toxicity. VDAC1 is the most abundant pore-forming protein of the outer mitochondrial membrane and, as a channel, it plays a pivotal role in regulating cellular bioenergetics, allowing the continuous exchange of ions and metabolites (ATP/ADP, Krebs cycle intermediates) between cytosol and mitochondria. In light of this evidence, we looked into the effects of Aβ oligomers on VDAC1 functions through electrophysiological and respirometric techniques. Our findings indicate that Aβ oligomers significantly modify the conductance, voltage dependency, and kinetic features of VDAC1, as well as its slight selectivity for anions, leading to a marked preference for cations. Given that VDAC1 is mainly involved in the trafficking of charged molecules in and out of mitochondria, a general reduction of cell viability and mitochondrial respiration was detected in neuroblastoma cells and primary cortical neurons exposed to Aβ oligomers. Interestingly, the toxic effect mediated by Aβ oligomers was counteracted by the use of NHK1, a small synthetic, cell-penetrating peptide that binds and modulates VDAC1. On these results, VDAC1 emerges as a crucial molecule in mitochondrial dysfunction in AD and as a promising pharmacological target for the development of new therapeutic avenues for this devastating neurodegenerative disease still without a cure.
Collapse
Affiliation(s)
- Fabrizio Cavallaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stefano Conti Nibali
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Pietro Caruso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stefania Zimbone
- Institute of Crystallography, National Research Council (CNR-IC), Catania, Italy
| | - Iolanda Rita Infantino
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Simona Reina
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Vito De Pinto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Angela Messina
- Department of Biological, Geological, Environmental Sciences, University of Catania, Catania, Italy
| | | | - Andrea Magrì
- Department of Biological, Geological, Environmental Sciences, University of Catania, Catania, Italy
| |
Collapse
|
55
|
Ghorbaninia M, Doroudgar S, Ganjalikhany MR. Delving into the crucial role of the initial structure in the dynamic and self-assembly of amyloid beta. Biochem Biophys Res Commun 2025; 758:151652. [PMID: 40117973 DOI: 10.1016/j.bbrc.2025.151652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
Alzheimer's disease involves the accumulation of amyloid beta (Aβ) monomers that form oligomers and fibrils in the brain. Studying the Aβ monomer is critical for understanding Aβ assembly and peptide behavior and has implications for drug design. Choosing a starting structure with a higher aggregation tendency for cost-effective MD studies and drug design is crucial. Previous studies have utilized distinct initial conformations, leading to varying results. Hence, this study was conducted to compare different initial conformations using the same MD simulation protocol to investigate the behavior and oligomerization propensity of different starting structures of Aβ during 1μs. The behavior of the monomers and their self-assembly systems were studied thoroughly, and the results revealed that highly helical Aβ monomers which used as starting structures retain high helix content during the simulation, and their tautomerization states did not cause significant changes in the structure. On the other hand, the Aβ extended and S-shaped monomers displayed the fingerprints of the fibril structure, which is believed to be more favorable for self-assembly. Self-assembly behaviors were seen for three S-shaped and three Aβ extended peptides. However, both conformations did not show stable β-sheet intermolecular interaction. For the Aβ16-22 monomer as a fragment of the Aβ that can assemble into fibrils, the impacts of capping and uncapping on the initial structure were also investigated. The results displayed that capped and uncapped structures can form oligomers with β-sheet at termini. However, in the capped state, β-sheet interactions were more stable and remained relatively longer than uncapped.
Collapse
Affiliation(s)
- Maryam Ghorbaninia
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Shirin Doroudgar
- Department of Internal Medicine and the Translational Cardiovascular Research Center, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, United States
| | - Mohamad Reza Ganjalikhany
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
56
|
Mishra R, Upadhyay A. An update on mammalian and non-mammalian animal models for biomarker development in neurodegenerative disorders. Cell Mol Life Sci 2025; 82:147. [PMID: 40192808 PMCID: PMC11977071 DOI: 10.1007/s00018-025-05668-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Neurodegeneration is one of the leading factor for death globally, affecting millions of people. Developing animal models are critical to understand biological processes and comprehend pathological hallmarks of neurodegenerative diseases. For decades, many animal models have served as excellent tools to determine the disease progression, develop diagnostic methods and design novel therapies against distinct pathologies. Here, we provide a comprehensive overview of both, mammalian and non-mammalian animal models, with a focus on three most common and aggressive neurodegenerative disorders: Alzheimer's disease, Parkinson's disease and Spinocerebellar ataxia-1. We highlight various approaches including transgene, gene transfer, and chemically-induced methods used to develop disease models. In particular, we discuss applications of both non-mammalian and mammalian contributions in research on neurodegeneration. It is exciting to learn the roles of animal models in disease pathomechanisms, identifying biomarkers and hence devising novel interventions to treat neuropathological conditions.
Collapse
Affiliation(s)
- Ribhav Mishra
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Chhattisgarh, 491002, India
| |
Collapse
|
57
|
Khan MS, Qureshi N, Khan R, Son YO, Maqbool T. CRISPR/Cas9-Based therapeutics as a promising strategy for management of Alzheimer's disease: progress and prospects. Front Cell Neurosci 2025; 19:1578138. [PMID: 40260080 PMCID: PMC12009953 DOI: 10.3389/fncel.2025.1578138] [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: 02/17/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025] Open
Abstract
CRISPR/Cas9 technology has revolutionized genetic and biomedical research in recent years. It enables editing and modulation of gene function with an unparalleled precision and effectiveness. Among the various applications and prospects of this technology, the opportunities it offers in unraveling the molecular underpinnings of a myriad of central nervous system diseases, including neurodegenerative disorders, psychiatric conditions, and developmental abnormalities, are unprecedented. In this review, we highlight the applications of CRISPR/Cas9-based therapeutics as a promising strategy for management of Alzheimer's disease and transformative impact of this technology on AD research. Further, we emphasize the role of CRISPR/Cas9 in generating accurate AD models for identification of novel therapeutic targets, besides the role of CRISPR-based therapies aimed at correcting AD-associated mutations and modulating the neurodegenerative processes. Furthermore, various delivery systems are reviewed and potential of the non-viral nanotechnology-based carriers for overcoming the critical limitations of effective delivery systems for CRISPR/Cas9 is discussed. Overall, this review highlights the promise and prospects of CRISPR/Cas9 technology for unraveling the intricate molecular processes underlying the development of AD, discusses its limitations, ethical concerns and several challenges including efficient delivery across the BBB, ensuring specificity, avoiding off-target effects. This article can be helpful in better understanding the applications of CRISPR/Cas9 based therapeutic approaches and the way forward utilizing enormous potential of this technology in targeted, gene-specific treatments that could change the trajectory of this debilitating and incurable illness.
Collapse
Affiliation(s)
- Mohamad Sultan Khan
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| | - Nousheen Qureshi
- Department of Higher Education, Government of Jammu and Kashmir, Srinagar, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Mohali, Punjab, India
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences and Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, Republic of Korea
| | - Tariq Maqbool
- Laboratory of Nanotherapeutics and Regenerative Medicine, Department of Nanotechnology, University of Kashmir, Srinagar, India
| |
Collapse
|
58
|
Armbrust F, Bickenbach K, Koudelka T, Joos C, Keller M, Tholey A, Pietrzik CU, Becker-Pauly C. HYTANE-Identified Latrophilin-3 Cleavage by Meprin β Leads to Loss of the Interaction Domains. J Proteome Res 2025; 24:1832-1844. [PMID: 40135725 PMCID: PMC11976865 DOI: 10.1021/acs.jproteome.4c00912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
The metalloprotease meprin β is upregulated in neurons and astrocytes of Alzheimer's disease patients' brains. While the role of meprin β as the β-secretase of amyloid precursor protein (APP) has been characterized, its broader substrate profile within the brain remains largely unexplored. Hence, to identify additional substrates, we conducted N-terminomics of brain lysates from mice overexpressing meprin β in astrocytes employing the Hydrophobic Tagging-Assisted N-terminal Enrichment (HYTANE) strategy. We observed 3906 (82.2%) N-terminal peptides and identified seven new substrates that match meprin β in terms of localization and cleavage specificity. Of note, the meprin β overexpressing mice show mild cognitive impairments caused by amyloidogenic APP processing alongside hyperactivity and altered exploratory behavior seemingly independent of APP cleavage. Hence, latrophilin-3 was of particular interest, as latrophilin-3 defects are associated with hyperactivity in mice and human. In brain lysates from mice overexpressing meprin β in astrocytes as well as in cellulo, we validated the cleavage of latrophilin-3, resulting in the release of two N-terminal domains. These domains promote interactions with neuronal proteins such as fibronectin leucine-rich repeat transmembrane proteins, promoting adequate synapse formation. Thus, meprin β might affect synaptic integrity by cleaving interaction domains of latrophilin-3, potentially exacerbating the observed hyperactivity phenotype.
Collapse
Affiliation(s)
- Fred Armbrust
- Biochemical
Institute, Unit for Degradomics of the Protease Web, University of Kiel, 24118 Kiel, Germany
| | - Kira Bickenbach
- Biochemical
Institute, Unit for Degradomics of the Protease Web, University of Kiel, 24118 Kiel, Germany
| | - Tomas Koudelka
- Systematic
Proteomics & Bioanalytics, Institute for Experimental Medicine, University of Kiel, 24105 Kiel, Germany
| | - Corentin Joos
- Biochemical
Institute, Unit for Degradomics of the Protease Web, University of Kiel, 24118 Kiel, Germany
| | - Maximilian Keller
- Institute
for Pathobiochemistry, University Medical
Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Andreas Tholey
- Systematic
Proteomics & Bioanalytics, Institute for Experimental Medicine, University of Kiel, 24105 Kiel, Germany
| | - Claus U. Pietrzik
- Institute
for Pathobiochemistry, University Medical
Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Christoph Becker-Pauly
- Biochemical
Institute, Unit for Degradomics of the Protease Web, University of Kiel, 24118 Kiel, Germany
| |
Collapse
|
59
|
Marsland P, Vore AS, Lutzke A, Gano A, Fischer A, Trapp S, Savage LM, Deak T. Sex-specific effects of chronic alcohol consumption across the lifespan in the transgenic Alzheimer's Disease (TgF344-AD) rat model. Brain Behav Immun 2025; 128:192-207. [PMID: 40187669 DOI: 10.1016/j.bbi.2025.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025] Open
Abstract
Alcohol consumption across the lifespan contributes to mood fluctuations and cognitive dysfunction, two neurobehavioral features also associated with Alzheimer's Disease and Related Dementias (ADRD). Yet, few studies have used rodent models to determine how a history of ethanol consumption across the lifespan might contribute to neurobehavioral and neuropathological features of ADRD. We exposed Wild Type (WT) and transgenic Fischer 344 CE rats (TgF344-AD) that have been genetically modified to express the human Amyloid Precursor Protein (APP) and presenilin-1 genes with mutations, to ethanol using a chronic, intermittent ethanol consumption model. Beginning at P28, rats were given a single bottle 10 % ethanol solution for 2 consecutive days, followed by 2 days of tap water. This pattern (2 days on, days off) was repeated for a total of 12 cycles until rats reached the age of ∼ 3 months, and repeated at 6 (Exp 1 and Exp 2) and 9 months of age (Exp 2). In experiment 1, ethanol consumption decreased alternations in a spontaneous alternation task in females, only at the 3-month time point, whereas TgF344-AD females showed increased contextual fear conditioning in the test of retention and reinstatement tests at 6 months of age. In experiment 2, a battery of anxiety-like behaviors (Elevated Plus Maze, Marble Burying, and Novelty Induced Hypophagia) were assessed following a 2-week abstinence period at 3, 6, and 9 months of age in ethanol-consuming rats. Data from the EPM and marble burying tasks revealed evidence of heightened anxiety-like behavior in Tg-F344-AD rats that varied by sex and age, with no significant effects of ethanol. In the novelty-induced hypophagia task, males with a history of ethanol consumption had a lower latency to approach a familiar, salient reward at 3 months old, but effects of ethanol were overall minimal. Examination of dorsal hippocampal gene expression at 6 months of age under basal conditions also revealed predominantly genotype and sex-specific effects on inflammation- and AD-related genes (App, Il-6, Bace1, Rage, Lrp-1). When examined at 9 months old following LPS challenge, ethanol increased inflammatory genes in males (Il-1β, Il-6) in the hippocampus, whereas ethanol decreased several inflammatory and AD-related genes (Hmgb1, Rage, Bace1, Lrp-1) in TgF344-AD females. Overall, these data provide further evidence that females are especially vulnerable to AD, and that a history of ethanol consumption had selective, rather than global, effects on AD- and inflammation-related genes following an inflammatory stimulus.
Collapse
Affiliation(s)
- Paige Marsland
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Andrew S Vore
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Ashley Lutzke
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Anny Gano
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Abigail Fischer
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Sarah Trapp
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Lisa M Savage
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States.
| |
Collapse
|
60
|
Gondelaud F, Bignon C, Ptchelkine D, Carrière F, Longhi S. A conserved motif in Henipavirus P/V/W proteins drives the fibrillation of the W protein from Hendra virus. Protein Sci 2025; 34:e70085. [PMID: 40100133 PMCID: PMC11917119 DOI: 10.1002/pro.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 03/20/2025]
Abstract
The Hendra (HeV) and Nipah (NiV) viruses are high-priority, biosafety level-4 pathogens that cause fatal neurological and respiratory disease. Their P gene encodes not only the P protein, an essential polymerase cofactor, but also the virulence factors V and W. We previously showed that the W protein of HeV (WHeV) forms amyloid-like fibrils and that one of its subdomains, PNT3, fibrillates in isolation. However, the fibrillation kinetics is much faster in the case of the full-length WHeV compared to PNT3, suggesting that another WHeV region contributes to the fibrillation process. In this work, we identified the region spanning residues 2-110 (PNT1) as the crucial region implicated in WHeV fibrillation. Through site-directed mutagenesis, combined with thioflavin T binding experiments and negative-staining transmission electron microscopy, we showed that a predicted cryptic amyloidogenic region (CAR) within PNT1 is the main driver of fibrillation and deciphered the underlying molecular mechanism. Using FTIR, we showed that PNT1 fibrils are enriched in cross β-sheets. Sequence alignment revealed conservation of the CAR across the Henipavirus genus and enabled the identification of a hitherto never reported pro-amyloidogenic motif. The ability to form fibrils was experimentally shown to be a common property shared by Henipavirus PNT1 proteins. Overall, this study sheds light on the molecular mechanisms underlying WHeV fibrillation and calls for future studies aimed at exploring the relevance of the newly identified pro-amyloidogenic motif as a valuable target for antiviral approaches.
Collapse
Affiliation(s)
- Frank Gondelaud
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257Aix Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Christophe Bignon
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257Aix Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Denis Ptchelkine
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257Aix Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| | - Frédéric Carrière
- Aix Marseille Univ, CNRS, UMR7281 Bioénergétique et Ingénierie des ProtéinesMarseilleFrance
| | - Sonia Longhi
- Laboratoire Architecture et Fonction des Macromolécules Biologiques (AFMB), UMR 7257Aix Marseille University and Centre National de la Recherche Scientifique (CNRS)MarseilleFrance
| |
Collapse
|
61
|
Malan A, Choudhary M, Kaur Bamrah P, Kumari D. Potential benefits of marine-derived compounds for slowing the advancement of Alzheimer's disease. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2025; 27:479-504. [PMID: 39373659 DOI: 10.1080/10286020.2024.2409869] [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: 03/05/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
The incidence of Alzheimer's is increasing and poses a significant social and economic burden. The pathogenesis involved in the expansion of AD includes neuronal oxidative damage, tau phosphorylation, amyloid beta aggregation, neuroinflammation, etc. Despite enormous efforts, there is currently no effective treatment or cure for this condition in the allopathic system. Marine compounds are appealing options and have a strong neuroprotective impact. Marine-derived compounds from sponges, algae, and marine invertebrates can be used for neuroprotection, with fewer adverse effects than synthetic drugs. Various compounds such as bryostatin-1, docosahexaenoic acid, spirolides, and astaxanthin, GV-971, have demonstrated outstanding activity and bioavailability.
Collapse
Affiliation(s)
- Aditya Malan
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, India
| | - Manjusha Choudhary
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, India
| | - Prabhjeet Kaur Bamrah
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, India
| | - Dipender Kumari
- Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra, 136119, India
| |
Collapse
|
62
|
Abbas K, Mustafa M, Alam M, Habib S, Ahmad W, Adnan M, Hassan MI, Usmani N. Multi-target approach to Alzheimer's disease prevention and treatment: antioxidant, anti-inflammatory, and amyloid- modulating mechanisms. Neurogenetics 2025; 26:39. [PMID: 40167826 DOI: 10.1007/s10048-025-00821-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) plaque accumulation, neurofibrillary tangles, neuroinflammation, and progressive cognitive decline, posing a significant global health challenge. Growing evidence suggests that dietary polyphenols may reduce the risk and progression of AD through multifaceted neuroprotective mechanisms. Polyphenols regulate amyloid proteostasis by inhibiting β/γ-secretase activity, preventing Aβ aggregation, and enhancing clearance pathways. Their strong antioxidant properties neutralize reactive oxygen species, chelate redox-active metals, and activate cytoprotective enzymes via Nrf2 signaling. This review examines the potential therapeutic targets, signaling pathways, and molecular mechanisms by which dietary polyphenols exert neuroprotective effects in AD, focusing on their roles in modulating amyloid proteostasis, oxidative stress, neuroinflammation, and cerebrovascular health. Polyphenols mitigate neuroinflammation by suppressing NF-κB signaling and upregulating brain-derived neurotrophic factor, supporting neuroplasticity and neurogenesis. They also enhance cerebrovascular health by improving cerebral blood flow, maintaining blood-brain barrier integrity, and modulating angiogenesis. This review examines the molecular and cellular pathways through which polyphenols exert neuroprotective effects, focusing on their antioxidant, anti-inflammatory, and amyloid-modulating roles. We also discuss their influence on key AD pathologies, including Aβ deposition, tau hyperphosphorylation, oxidative stress, and neuroinflammation. Insights from clinical and preclinical studies highlight the potential of polyphenols in preventing or slowing AD progression. Future research should explore personalized dietary strategies that integrate genetic and lifestyle factors to optimize the neuroprotective effects of polyphenols.
Collapse
Affiliation(s)
- Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Waleem Ahmad
- Department of Medicine, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'Il, Ha'il, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Nazura Usmani
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
63
|
Fathalla M, Abolibda TZ, Al-Kaff NS, Rudolf R, Sarkar B, Albarbarawi O, Gomha SM, Said MA. Porphyrin Schiff bases as potential Alzheimer's disease drug candidates: Synthesis, in silico and DFT studies. J Mol Struct 2025; 1327:141251. [DOI: 10.1016/j.molstruc.2024.141251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
|
64
|
Kuznetsov AV. Evaluating the Combined Neurotoxicity of Amyloid Beta and Tau Oligomers in Alzheimer's Disease: A Novel Cellular-Level Criterion. J Biomech Eng 2025; 147:041003. [PMID: 39840747 DOI: 10.1115/1.4067701] [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: 11/03/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
A criterion characterizing the combined neurotoxicity of amyloid beta and tau oligomers is suggested. A mathematical model for calculating the value of this criterion during senile plaque and neurofibrillary tangle (NFT) formation is proposed. Computations show that for physiologically relevant parameter values, the value of the criterion increases approximately linearly with time. Once neurofibrillary tangles begin forming in addition to senile plaques, there is an increase in the slope characterizing the rate at which the criterion increases. The critical value of the criterion at which a neuron dies is estimated. Unless the production rates of amyloid beta and tau monomers are very large, computations predict that for the accumulated toxicity to reach the critical value, the neural machinery responsible for the degradation of amyloid beta and tau monomers and aggregates must become dysfunctional. The value of the criterion after 20 years of the aggregation process is strongly influenced by the deposition rates of amyloid beta and tau oligomers into senile plaques and NFTs. This suggests that deposition of amyloid beta and tau oligomers into senile plaques and NFTs may reduce accumulated toxicity by sequestering more toxic oligomeric species into less toxic insoluble aggregates.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695-7910
| |
Collapse
|
65
|
Liu Y, Liu Y, Shi L, Zhang X, Liu K, He S. Selenium ameliorates cognitive impairment through activating BDNF/TrkB pathway. J Trace Elem Med Biol 2025; 88:127599. [PMID: 39837256 DOI: 10.1016/j.jtemb.2025.127599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder that primarily affects older adults. Selenium, an essential micronutrient for humans, plays a crucial role in the body's normal physiological and metabolic processes. A long-term deficiency in selenium intake can lead to various diseases and even contribute to the ageing process. This study aims to explore the ameliorative effect of selenium on cognitive impairment in 3 × Tg-AD mice and to determine if its effects are related to the BDNF/TrkB pathway. METHODS We employed the APP/PS1/tau 3 × Tg-AD mouse model for dietary selenium intervention. Behavioural experiments were conducted to assess learning and memory. Additionally, we measured selenium and GSH-Px levels in whole blood and brain tissue. Neuronal apoptosis in the hippocampus was observed using transmission electron microscopy. The expressions of Aβ, P-tau, BDNF, TrkB, and CREB were measured via RT-qPCR, while the expressions of Aβ, P-tau, BDNF, TrkB, p-CREB, and CREB were quantified using Western blot analysis. RESULTS Our findings indicate that selenium supplementation can improve spatial learning and memory deficiencies in 3 × Tg-AD mice. Selenium supplementation increased selenium and GSH-Px levels in the brain tissue of 3 × Tg-AD mice and significantly enhanced neuronal conditions. Furthermore, the expression levels of proteins related to the BDNF/TrkB pathway significantly increased following selenium supplementation. CONCLUSIONS Our study demonstrates that selenium can ameliorate memory impairment in 3 × Tg-AD mice by activating the BDNF/TrkB pathway.
Collapse
Affiliation(s)
- Yu Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan 750004, China; Department of Medical Administration, Baotou Central Hospital, Baotou, Inner Mongolia 014040, China
| | - Ye Liu
- Infectious Diseases Clinic, The Fourth People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750021, China
| | - Liping Shi
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan 750004, China
| | - Xue Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan 750004, China
| | - Kunmei Liu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Incubation Base of the National Key Laboratory, Ningxia Medical University, Yinchuan 750004, China.
| | - Shulan He
- Department of Epidemiology and Health Statistics, School of Public Health, Ningxia Medical University, Yinchuan 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, Yinchuan 750004, China.
| |
Collapse
|
66
|
Raj V, Raorane CJ, Shastri D, Kim JH, Lee S. Sulfonic acid functionalized β-amyloid peptide aggregation inhibitors and antioxidant agents for the treatment of Alzheimer's disease: Combining machine learning, computational, in vitro and in vivo approaches. Int J Biol Macromol 2025; 299:140142. [PMID: 39842570 DOI: 10.1016/j.ijbiomac.2025.140142] [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: 05/21/2024] [Revised: 11/01/2024] [Accepted: 01/20/2025] [Indexed: 01/24/2025]
Abstract
Alzheimer's disease (AD) is characterized as a neurodegenerative disorder that is caused by plaque formation by accumulating β-amyloid (Aβ), leading to neurocognitive function and impaired mental development. Thus, targeting Aβ represents a promising target for the development of therapeutics in AD management. Several functionalized sulfonic acid molecules have been reported, including tramiprosate prodrug, which is currently in clinical trial III and exhibits a good response in mild to moderate AD patients. Therefore, expanding upon this approach, we hypothesized that the sulfonic acid functionalized aromatic class molecule might demonstrate a good inhibitory effect against β-amyloid aggregation, leading to a decrease in the progression burden of AD. We used computational and in vitro approaches to establish effective compounds. As a result, three potent hit molecules were selected based on binding score as well as availability. In the case of safety profile of compounds, in vitro using human neuroblastoma SH-SY5Y cells and in vivo using C. elegans was performed at doses up to 500 μM; no difference in viability was exhibited between control and treatment groups. However, H2O2-induced ROS stress was significantly reduced in neuroblastoma cells after treatment. The AFM and ThT-embedded β-amyloid1-42 kinetic studies confirmed B-PEA-MBSA and H-HPA-NSA potency. H-HPA-NSA arrested elongation phase of Aβ aggregation in kinetic study at a lower concentration (10 μM), while B-PEA-MBSA reduced the intensity of stationary phase at a dose of 100 μM. Thus, based on the outcomes, it can be suggested that B-PEA-MBSA and H-HPA-NSA can prevent β-amyloid aggregation with mild to moderate AD.
Collapse
Affiliation(s)
- Vinit Raj
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea
| | | | - Divya Shastri
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea; College of Pharmacy, Keimyung University, 1095 Dalgubeol-daero, Dalseo-Gu, Daegu 42601, Republic of Korea
| | - Jae Hyun Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea
| | - Sangkil Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea.
| |
Collapse
|
67
|
Zhang Z, Li R, Zhou Y, Huang S, Hou Y, Pei G. Dietary Flavonoid Chrysin Functions as a Dual Modulator to Attenuate Amyloid-β and Tau Pathology in the Models of Alzheimer's Disease. Mol Neurobiol 2025; 62:4274-4291. [PMID: 39432184 DOI: 10.1007/s12035-024-04557-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
Growing evidence indicates that healthy diets are associated with a slower progression of Alzheimer's disease (AD). Flavonoids are among the most abundant natural products in diets beneficial to AD, such as the Mediterranean diet. However, the effect and mechanism of these dietary flavonoids on AD remains incompletely understood. Here, we found that a representative dietary natural flavonoid, chrysin (Chr), significantly ameliorated cognitive impairment and AD pathology in APP/PS1 mice. Furthermore, mechanistic studies showed that Chr significantly reduced the levels of amyloid-β (Aβ) and phosphorylated tau (p-tau), along with dual inhibitory activity against β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and glycogen synthase kinase 3β (GSK3β). Moreover, the effect of Chr was further confirmed by EW233, a structural analog of Chr that exhibited an improved pharmacokinetic profile. To further verify the role of Chr and EW233, we utilized our previously established chimeric human cerebral organoid (chCO) model for AD, in which astrogenesis was promoted to mimic the neuron-astrocyte ratio in human brain tissue, and similar dual inhibition of Aβ and p-tau was also observed. Altogether, our study not only reveals the molecular mechanisms through which dietary flavonoids, such as Chr, mitigate AD pathology, but also suggests that identifying a specific constituent that mimics some of the benefits of these healthy diets could serve as a promising approach to discover new treatments for AD.
Collapse
Affiliation(s)
- Zhen Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Rongyao Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shichao Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yujun Hou
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
68
|
Petersen RC, Graf A, Atkins AS, Brys M, Murphy J, Miller DS, Reyderman L, Siemers E, Smith J, Carrillo MC, Weber CJ. Understanding the impact of amyloid beta targeted therapies on biomarkers and clinical endpoints in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2025; 11:e70069. [PMID: 40417267 PMCID: PMC12102660 DOI: 10.1002/trc2.70069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 05/27/2025]
Abstract
The Alzheimer's disease (AD) scientific field continues to make significant advances in early detection and treatments, which importantly rest on advances in our fundamental understanding of AD pathobiology and its contribution to cognitive decline. Clinical readouts of monoclonal antibodies against various forms of the amyloid beta (Aβ) protein indicate that the impact of these treatments may extend beyond reduction in amyloid plaques. The Alzheimer's Association Research Roundtable meeting held on May 17 and 18, 2022, reviewed our understanding to date of the impact of treatments targeting various species of Aβ; its impact on other related pathophysiology including tau; and ultimately, its effects on neurodegeneration and clinical decline, driven by the latest available data. Participants discussed the current evidence for a causal relationship among amyloid accumulation, tau alteration, and cognitive decline; the effect of anti-amyloid therapies on clinical and biomarker endpoints; and how we can accelerate the pathway to therapeutic approval and what should guide us for the near future. Highlights The Alzheimer's Association Research Roundtable convened leaders from industry and academia, as well as patients, clinicians, and government and regulatory agency scientists to discuss the topic "Current Understanding of AD Pathophysiology & Impact of Amyloid-beta Targeted Treatments on Biomarkers and Clinical Endpoints."The totality of scientific evidence (clinical trials, animal data, modeling, and observational studies) on the relationship between amyloid beta (Aβ), amyloid, tau, and cognitive impairment is helping our understanding of the downstream effects and overall importance of lowering amyloid plaque load.Based on data from multiple phase 2 and 3 clinical trials of anti-amyloid monoclonal antibodies, there is strong evidence to support that a sufficiently large reduction in amyloid plaque load to near-normal levels is associated with positive changes in tau biomarkers and clinical endpoints.Reduction of Aβ plaque, measured easily by plasma amyloid biomarkers, is reasonably likely to predict benefit in clinical outcome measures.
Collapse
Affiliation(s)
| | - Ana Graf
- Novartis Pharma AGBaselSwitzerland
| | | | | | | | | | | | - Eric Siemers
- Acumen PharmaceuticalsCharlottesvilleVirginiaUSA
| | | | | | | |
Collapse
|
69
|
Zhang M, Xu Y, Sun X, Shi X, Liang H, Chen X, Cui W, Fan Y, Ma J, Wang H. Pharmacokinetics, Tissue Distribution, and Excretion of 9-Methylfascaplysin, a Potential Anti-Alzheimer's Disease Agent. Electrophoresis 2025; 46:452-461. [PMID: 40099706 DOI: 10.1002/elps.8135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/14/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
9-Methylfascaplysin, a derivative of the marine natural product fascaplysin, has shown promising anti- Alzheimer's disease (AD) potential through its anti-β-amyloid (Aβ) neuroprotective effects. However, the pharmacokinetics (PK) of 9-methylfascaplysin, crucial for its preclinical evaluation, have not been thoroughly studied. In this study, we developed and validated a sensitive and accurate ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method for the quantification of 9-methylfascaplysin in rat plasma. The method demonstrated a lower limit of detection (LLOD) of 1 ng/mL and a linear quantification range of 5-2000 ng/mL. The PK study in rat plasma was conducted. After intragastric administration, the plasma concentration of 9-methylfascaplysin peaked at a maximum concentration (Cmax) of 193.4 ng/m and an enterohepatic circulation (EHC) phenomenon was observed. By comparing the area under the plasma concentration-time curve (AUC) values obtained from intragastric and intravenous administrations, the absolute oral bioavailability (F) of 9-methylfascaplysin was determined as 18.3%. The tissue distribution study revealed that following a single intragastric administration, 9-methylfascaplysin was most concentrated in the stomach, followed by the small intestine, large intestine, liver, kidney, brain, lung, spleen, and heart in descending order. Furthermore, the excretion profiles of 9-methylfascaplysin in rat urine and feces were studied. The results of this study provide valuable insights into the PK behavior of 9-methylfascaplysin and serve as a foundation for its further preclinical evaluation and potential clinical application as an anti-AD agent.
Collapse
Affiliation(s)
- Manman Zhang
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yu Xu
- Key Laboratory of Drug Monitoring and Control of Zhejiang Province, National Narcotics Laboratory Zhejiang Regional Center, Hangzhou, China
| | - Xingjian Sun
- School of Mathematical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaolu Shi
- Key Laboratory of Drug Monitoring and Control of Zhejiang Province, National Narcotics Laboratory Zhejiang Regional Center, Hangzhou, China
| | - Hongze Liang
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
| | - Xiaowei Chen
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Wei Cui
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Yilei Fan
- The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou, China
| | - Jianfeng Ma
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haixing Wang
- Key Laboratory of Drug Monitoring and Control of Zhejiang Province, National Narcotics Laboratory Zhejiang Regional Center, Hangzhou, China
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
70
|
Zhu Y, Dai Y, Tian Y. The Peptide PROTAC Modality: A New Strategy for Drug Discovery. MedComm (Beijing) 2025; 6:e70133. [PMID: 40135198 PMCID: PMC11933449 DOI: 10.1002/mco2.70133] [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: 07/04/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
In recent years, proteolysis targeting chimera (PROTAC) technology has made significant progress in the field of drug development. Traditional drugs mainly focus on inhibiting or activating specific proteins, while PROTAC technology provides new ideas for treating various diseases by inducing the degradation of target proteins. Especially for peptide PROTACs, due to their unique structural and functional characteristics, they have become a hot research topic. This review provides a detailed description of the key components, mechanisms, and design principles of peptide PROTACs, elaborates on their applications in skin-related diseases, oncology, and other potential therapeutic fields, analyzes their advantages and challenges, and looks forward to their future development prospects. The development of peptide PROTAC technology not only opens up new paths for drug research and development, but also provides new ideas for solving the resistance and safety issues faced by traditional small-molecule drugs. Compared with small-molecule PROTACs, peptide PROTACs have advantages such as multitargeting, biodegradability, low toxicity, and flexibility in structural design. With the deepening of research and the continuous maturity of technology, peptide PROTACs are expected to become one of the important strategies for future drug discovery, providing new hope for the treatment of more intractable diseases. Peptide PROTACs are ushering in a new era of precision medicine.
Collapse
Affiliation(s)
- Youmin Zhu
- Shanghai AZ Science and Technology Co., Ltd.ShanghaiChina
| | - Yu Dai
- Shanghai AZ Science and Technology Co., Ltd.ShanghaiChina
- School of BiotechnologyEast China University of Science and TechnologyShanghaiChina
| | - Yuncai Tian
- Shanghai AZ Science and Technology Co., Ltd.ShanghaiChina
| |
Collapse
|
71
|
Suloh H, Ojha SK, Kartawy M, Hamoudi W, Tripathi MK, Bazbaz W, Schottlender N, Ashery U, Khaliulin I, Amal H. Shared early molecular mechanisms revealed in P301S and 5xFAD Alzheimer's disease mouse models. Transl Psychiatry 2025; 15:97. [PMID: 40140365 PMCID: PMC11947184 DOI: 10.1038/s41398-025-03321-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by early molecular events that influence disease progression. Still, the molecular mechanisms caused by different mutations of AD are not understood. We have performed a multidisciplinary study to investigate and compare the early stages of the pathology in two transgenic AD mouse models: P301S and 5xFAD. Using SNOTRAP-based mass spectrometry, we assessed changes in S-nitrosylation, a nitric oxide-mediated post-translational modification, of proteins in both models during their juvenile age. The increased levels of 3-nitrotyrosine confirmed nitrosative stress in the mutant mice. Systems biology analysis revealed shared processes between the models, particularly in the γ-aminobutyric acid (GABA)ergic and glutamatergic neurotransmission processes. In the P301S model, we identified 273 S-nitrosylated (SNOed) proteins in the cortex, with 244 proteins uniquely SNOed in the diseased mice. In the 5xFAD model, 309 SNOed proteins were identified. We have found altered proteins expression of different glutamate/GABA-related markers in the cortex and hippocampus of both AD mouse models. Additionally, the phosphorylation levels of the mTOR signaling components revealed hyperactivation of this pathway in P301S mice. Conversely, 5xFAD mice showed no significant changes in mTOR signaling except for elevated phosphorylation of the ribosomal protein S6 in the cortex. Our findings revealed key molecular mechanisms in the two AD mouse models during their early stages. These mechanisms could serve as potential biomarkers and therapeutic targets for early-stage AD.
Collapse
Affiliation(s)
- Huda Suloh
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wisam Bazbaz
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nofar Schottlender
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts; Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
72
|
Tang H, Andrikopoulos N, Li Y, Ke S, Sun Y, Ding F, Ke PC. Emerging biophysical origins and pathogenic implications of amyloid oligomers. Nat Commun 2025; 16:2937. [PMID: 40133283 PMCID: PMC11937510 DOI: 10.1038/s41467-025-58335-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
The amyloid hypothesis has been a leading narrative concerning the pathophysiological foundation of Alzheimer's and Parkinson's disease. At the two ends of the hypothesis lie the functional protein monomers and the pathology-defining amyloid fibrils, while the early stages of protein aggregation are populated by polymorphic, transient and neurotoxic oligomers. As the structure and activity of oligomers are intertwined, here we show oligomers arising from liquid-liquid phase separation and β-barrel formation, their routes to neurodegeneration, and their role in cerebrovascular perturbation. Together, this Perspective converges on the multifaceted oligomer-axis central to the pathological origin and, hence, the treatment of amyloid diseases.
Collapse
Affiliation(s)
- Huayuan Tang
- Department of Engineering Mechanics, Hohai University, Nanjing, 211100, China
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA
| | - Nicholas Andrikopoulos
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Yuhuan Li
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Stone Ke
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, 30 Royal Parade, Parkville, VIC, 3052, Australia
| | - Yunxiang Sun
- School of Physical Science and Technology, Ningbo University, Ningbo, 315211, China.
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA.
| | - Pu Chun Ke
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
73
|
Zhou Q, Wang W, Deng C. Advancements in Proteolysis Targeting Chimeras for Targeted Therapeutic Strategies in Alzheimer's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04838-0. [PMID: 40133753 DOI: 10.1007/s12035-025-04838-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/09/2025] [Indexed: 03/27/2025]
Abstract
The presence of hyperphosphorylated Tau proteins, which mislocalize and form neurofibrillary tangles, and the accumulation of amyloid-β plaques are hallmark features of Alzheimer's disease (AD). These toxic protein aggregates contribute to synaptic impairment and neuronal dysfunction, underscoring the need for strategies aimed at effectively clearing or reducing these aggregates in the treatment of AD. In recent years, proteolysis targeting chimera (PROTAC) technology has emerged as a promising approach for selectively degrading dysfunctional proteins rather than merely inhibiting their function. This approach holds great potential for developing more effective interventions that could slow AD progression and improve patient outcomes. In this review, we first examine the pathological mechanisms underlying AD, focusing on abnormal protein degradation and accumulation. We then explore the evolution of PROTAC technology, its mechanisms of action, and the current status of drug development. Finally, we discuss the latest findings regarding the application of PROTACs in AD therapy, highlighting the potential benefits and limitations of this technology. Although promising, further clinical research is necessary to fully assess the safety and efficacy of PROTAC-based therapies for AD treatment.
Collapse
Affiliation(s)
- Qiuzhi Zhou
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weixia Wang
- School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing, 100191, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
74
|
Bashirova N, Schölzel F, Hornig D, Scheidt HA, Krueger M, Salvan G, Huster D, Matysik J, Alia A. The Effect of Polyethylene Terephthalate Nanoplastics on Amyloid-β Peptide Fibrillation. Molecules 2025; 30:1432. [PMID: 40286031 PMCID: PMC11990616 DOI: 10.3390/molecules30071432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Exposure of organisms to nanoplastics (NPs) is inevitable given their global abundance and environmental persistence. Polyethylene terephthalate (PET) is a common plastic used in a wide range of products, including clothing and food and beverage packaging. Recent studies suggest that NPs can cross the blood-brain barrier and cause potential neurotoxicity. It is widely known that aggregation of amyloid beta (Aβ) peptides in the brain is a pathological hallmark of Alzheimer's disease (AD). While the impact of nanoplastics such as polystyrene (PS) on amyloid aggregation has been studied, the effects of PET NPs remain unexplored. In this study, we examined the effect of PET NPs of different sizes (PET50nm and PET140nm) and concentrations (0, 10, 50, and 100 ppm) on the fibrillation of Aβ1-40. Our results showed that the presence of PET50nm as well as PET140nm decreased the lag phase of the fibrillation processes in a dose- and size-dependent manner from 6.7 ± 0.08 h for Aβ in the absence of PET (Aβcontrol) to 3.1 ± 0.03 h for PET50nm and 3.8 ± 0.06 h for PET140nm. CD spectroscopy showed that PET50nm significantly impacts the structural composition of Aβ aggregates. A significant rise in antiparallel β-sheet content and β-turn structure and a substantial reduction in other structures were observed in the presence of 100 ppm PET50nm. These changes indicate that higher concentrations (100 ppm) of PET50nm promote more rigid and uniform peptide aggregates. Although PET50nm NPs influence the kinetics of aggregation and secondary structure, the overall morphology of the resulting fibrils remains largely unaltered, as seen using transmission electron microscopy. Also, the local cross-β structure of the fibrils was not affected by the presence of PET50nm NPs during fibrillation, as confirmed using 13C solid-state NMR spectroscopy. Overall, these findings show that PET NPs accelerate amyloid fibril formation and alter the secondary structure of Aβ fibrils. These results also indicate that the accumulation of PET-NPs in the brain may facilitate the progression of various neurodegenerative diseases, including Alzheimer's disease.
Collapse
Affiliation(s)
- Narmin Bashirova
- Institute of Medical Physics and Biophysics, Leipzig University, D-04107 Leipzig, Germany (D.H.)
- Institute of Analytical Chemistry, Leipzig University, D-04103 Leipzig, Germany;
| | - Franziska Schölzel
- Institute of Physics, Chemnitz University of Technology, D-09126 Chemnitz, Germany (G.S.)
- Center for Materials, Architectures and Integration of Nanomembranes, Chemnitz University of Technology, D-09126 Chemnitz, Germany
| | - Dominik Hornig
- Center for Materials, Architectures and Integration of Nanomembranes, Chemnitz University of Technology, D-09126 Chemnitz, Germany
- Institute of Chemistry, Chemnitz University of Technology, D-09107 Chemnitz, Germany
| | - Holger A. Scheidt
- Institute of Medical Physics and Biophysics, Leipzig University, D-04107 Leipzig, Germany (D.H.)
| | - Martin Krueger
- Institute of Anatomy, Leipzig University, D-04107 Leipzig, Germany
| | - Georgeta Salvan
- Institute of Physics, Chemnitz University of Technology, D-09126 Chemnitz, Germany (G.S.)
- Institute of Chemistry, Chemnitz University of Technology, D-09107 Chemnitz, Germany
| | - Daniel Huster
- Institute of Medical Physics and Biophysics, Leipzig University, D-04107 Leipzig, Germany (D.H.)
| | - Joerg Matysik
- Institute of Analytical Chemistry, Leipzig University, D-04103 Leipzig, Germany;
| | - A. Alia
- Institute of Medical Physics and Biophysics, Leipzig University, D-04107 Leipzig, Germany (D.H.)
- Leiden Institute of Chemistry, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
75
|
Xia Y, Tsim KWK, Wang WX. Disruption of Copper Redox Balance and Dysfunction under In Vivo and In Vitro Alzheimer's Disease Models. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:238-249. [PMID: 40144323 PMCID: PMC11934196 DOI: 10.1021/envhealth.4c00175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 03/28/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder disease mainly caused by extracellular senile plaques (SP) formed by β-amyloid (Aβ1-42) protein deposits. Copper (Cu) is an essential metal involved in neural system, and its homeostasis is the key to maintain its proper function. Herein, the subcellular locations of Cu(I) and Cu(II) in human neurodegenerative disease SH-SY5Y cells and AD mouse brains were imaged. We found that the content of Cu(II) decreased while that of Cu(I) increased under Aβ exposure, which were further verified in the brain tissues of the AD mouse model, strongly suggesting the disruption of Cu homeostasis under Aβ exposure or AD. Remarkably, the mitochondrial and lysosomal Cu(II) decreased significantly, whereas Cu(I) decreased in mitochondria but increased in lysosome. Lysosomes digested the damaged mitochondria via mitophagy to remove excess Cu(I) and maintain Cu homeostasis. The Aβ induced Cu(I) in mitochondria resulted in an overformation of reactive oxygen species and altered the morphology of this organelle. Due to the oxidative stress, glutathione (GSH) was converted into glutathione disulfide (GSSG), and Cu(I) bound with GSH was further released into the cytoplasm and absorbed by the lysosome. Transcriptomic analysis showed that genes (ATP7A/B) related to Cu transportation were upregulated, whereas genes related to mitochondrial complex were down-regulated, representing the damage of this organelle. This study demonstrated that Aβ exposure caused the disruption of intracellular homeostasis by reducing Cu(II) to Cu(I) and damaging the mitochondria, which further triggered detoxification by the lysosome. Our finding provided new insights in Aβ and AD induced Cu redox transformation and toxicity.
Collapse
Affiliation(s)
- Yiteng Xia
- School
of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong
Kong, China
- Research
Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| | - Karl W. K. Tsim
- Division
of Life Science, Hong Kong University of
Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Wen-Xiong Wang
- School
of Energy and Environment and State Key Laboratory of Marine Pollution, City University of Hong Kong, Kowloon, Hong
Kong, China
- Research
Centre for the Oceans and Human Health, City University of Hong Kong Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
76
|
Dindar Z, Anbaraki A, Hosseini SS, Harati Z, Bahrami A, Balalaie S, Ghobeh M, Mahdavi M, Seyedarabi A. The Use of Natural Volatile Compounds on the Fibrillation Domain of Amyloid Beta (GSNKGAIIGLM)─Towards Promising Agents to Combat Alzheimer's Disease. ACS Chem Neurosci 2025; 16:1086-1102. [PMID: 40059298 DOI: 10.1021/acschemneuro.4c00768] [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: 03/20/2025] Open
Abstract
Alzheimer's disease (AD), which is caused by the accumulation of amyloid-beta, is a major medical concern today. Controlling these aggregates is critical to drug development, but delivering them effectively into the bloodstream poses significant challenges. In this context, aromatherapy has been proposed as an innovative and promising approach for AD disease. The volatile compounds cinnamaldehyde, phenylethyl alcohol, α-asarone, and β-caryophyllene have neuroprotective effects that can be effective in the treatment of neurodegenerative diseases like AD. The amyloid-beta (Aβ) fragment (25-35), which retains the properties of the full-length Aβ is used as a suitable model to evaluate the potential toxicity associated with AD. This study investigated the effects of the four mentioned volatile compounds at four different concentrations on the fibrillation process of the Aβ (25-35) peptide. Structural changes in the peptide have been analyzed using various techniques such as fluorescence probing, far-UV circular dichroism spectroscopy (CD), and atomic force microscopy (AFM). Fluorescence probing results showed that these compounds can effectively prevent the formation of amyloid fibrils by forming chemical bonds with the intermediate species. CD spectroscopy results indicated a decrease in β-sheet content of fibrils and confirmed the effect of pH on structural changes. AFM analysis revealed that volatile compounds effectively prevented the formation of amyloid fibrils at different concentrations and changed the average size of intermediates and oligomeric species. These findings show a promising future for AD patients and emphasize the importance of natural compounds in the treatment and prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zahra Dindar
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384 Tehran, Iran
| | - Afrooz Anbaraki
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384 Tehran, Iran
| | - Seyyed Sina Hosseini
- Peptide Chemistry Research Institute, K. N. Toosi University of Technology, P.O. Box 15875-4416 Tehran, Iran
| | - Zohreh Harati
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384 Tehran, Iran
| | - Aida Bahrami
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384 Tehran, Iran
| | - Saeed Balalaie
- Peptide Chemistry Research Institute, K. N. Toosi University of Technology, P.O. Box 15875-4416 Tehran, Iran
| | - Maryam Ghobeh
- Department of Biology, Science and Research Branch, Islamic Azad University, P.O. Box 1477893855 Tehran, Iran
| | - Majid Mahdavi
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384 Tehran, Iran
| | - Arefeh Seyedarabi
- Institute of Biochemistry and Biophysics, University of Tehran, P.O. Box 13145-1384 Tehran, Iran
| |
Collapse
|
77
|
Nashiro K, Cahn BR, Choi P, Lee HRJ, Satchi S, Min J, Yoo HJ, Cho C, Mercer N, Sordo L, Head E, Choupan J, Mather M. Daily mindfulness practice with and without slow breathing has opposing effects on plasma amyloid beta levels. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.10.25323695. [PMID: 40162262 PMCID: PMC11952629 DOI: 10.1101/2025.03.10.25323695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Prior research suggests that meditation may slow brain aging and reduce the risk of Alzheimer's disease (AD). However, we lack research systematically examining what aspect(s) of meditation may drive such benefits. In particular, it is unknown how breathing patterns during meditation might influence health outcomes associated with AD. In this study, we examined whether two types of mindfulness meditation practices - one with slow breathing and one with normal breathing - differently affect plasma amyloid beta (Aβ) relative to a no-intervention control group. One week of daily mindfulness practice with slow breathing decreased plasma Aβ levels whereas one week of daily mindfulness practice with normal breathing increased them. The no-intervention control group showed no changes in plasma Aβ levels. Slow breathing appears to be a factor through which meditative practices can influence pathways relevant for AD.
Collapse
|
78
|
Adam Wesołowski P, Yang B, Davolio AJ, Woods EJ, Pracht P, Bojarski KK, Wierbiłowicz K, Payne MC, Wales DJ. Decoding Solubility Signatures from Amyloid Monomer Energy Landscapes. J Chem Theory Comput 2025; 21:2736-2756. [PMID: 39988900 PMCID: PMC11912213 DOI: 10.1021/acs.jctc.4c01623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
This study investigates the energy landscapes of amyloid monomers, which are crucial for understanding protein misfolding mechanisms in Alzheimer's disease. While proteins possess inherent thermodynamic stability, environmental factors can induce deviations from native folding pathways, leading to misfolding and aggregation, phenomena closely linked to solubility. Using the UNOPTIM program, which integrates the UNRES potential into the Cambridge energy landscape framework, we conducted single-ended transition state searches and employed discrete path sampling to compute kinetic transition networks starting from PDB structures. These kinetic transition networks consist of local energy minima and the transition states that connect them, which quantify the energy landscapes of the amyloid monomers. We defined clusters within each landscape using energy thresholds and selected their lowest-energy structures for the structural analysis. Applying graph convolutional networks, we identified solubility trends and correlated them with structural features. Our findings identify specific minima with low solubility, characteristic of aggregation-prone states, highlighting the key residues that drive reduced solubility. Notably, the exposure of the hydrophobic residue Phe19 to the solvent triggers a structural collapse by disrupting the neighboring helix. Additionally, we investigated selected minima to determine the first passage times between states, thereby elucidating the kinetics of these energy landscapes. This comprehensive approach provides valuable insights into the thermodynamics and kinetics of Aβ monomers. By integration of multiple analytical techniques to explore the energy landscapes, our study investigates structural features associated with reduced solubility. These insights have the potential to inform future therapeutic strategies aimed at addressing protein misfolding and aggregation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Patryk Adam Wesołowski
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Bojun Yang
- Shenzhen
College of International Education, Antuoshan sixth Road, Shenzhen 518040, China
| | - Anthony J. Davolio
- Theory
of Condensed Matter Group, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, U.K.
| | - Esmae J. Woods
- Department
of Engineering, University of Cambridge, Trumpington Street, Cambridge CB2 1PZ, U.K.
| | - Philipp Pracht
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Krzysztof K. Bojarski
- Department
of Physical Chemistry, Gdansk University
of Technology, Narutowicza
11/12, Gdansk 80-233, Poland
| | - Krzysztof Wierbiłowicz
- Department
of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, 1335 Lee Street, Charlottesville, Virginia 22908, United States
| | - Mike C. Payne
- Theory
of Condensed Matter Group, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, U.K.
| | - David J. Wales
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| |
Collapse
|
79
|
Belbasis L, Morris S, van Duijn C, Bennett D, Walters R. Mendelian randomization identifies proteins involved in neurodegenerative diseases. Brain 2025:awaf018. [PMID: 40037332 DOI: 10.1093/brain/awaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 10/26/2024] [Accepted: 12/20/2024] [Indexed: 03/06/2025] Open
Abstract
Proteins are involved in multiple biological functions. High-throughput technologies have allowed the measurement of thousands of proteins in population biobanks. In this study, we aimed to identify proteins related to Alzheimer's disease, Parkinson's disease, multiple sclerosis and amyotrophic lateral sclerosis by leveraging large-scale genetic and proteomic data. We performed a two-sample cis Mendelian randomization study by selecting instrumental variables for the abundance of >2700 proteins measured by either Olink or SomaScan platforms in plasma from the UK Biobank and the deCODE Health Study. We also used the latest publicly available genome-wide association studies for the neurodegenerative diseases of interest. The potentially causal effect of proteins on neurodegenerative diseases was estimated based on the Wald ratio. We tested 13 377 protein-disease associations, identifying 169 associations that were statistically significant (5% false discovery rate). Evidence of co-localization between plasma protein abundance and disease risk (posterior probability > 0.80) was identified for 61 protein-disease pairs, leading to 50 unique protein-disease associations. Notably, 23 of 50 protein-disease associations corresponded to genetic loci not previously reported by genome-wide association studies. The two-sample Mendelian randomization and co-localization analysis also showed that APOE abundance in plasma was associated with three subcortical volumes (hippocampus, amygdala and nucleus accumbens) and white matter hyper-intensities, whereas PILRA and PILRB abundance in plasma was associated with caudate nucleus volume. Our study provided a comprehensive assessment of the effect of the human proteome that is currently measurable through two different platforms on neurodegenerative diseases. The newly associated proteins indicated the involvement of complement (C1S and C1R), microglia (SIRPA, SIGLEC9 and PRSS8) and lysosomes (CLN5) in Alzheimer's disease; the interleukin-6 pathway (CTF1) in Parkinson's disease; lysosomes (TPP1), blood-brain barrier integrity (MFAP2) and astrocytes (TNFSF13) in amyotrophic lateral sclerosis; and blood-brain barrier integrity (VEGFB), oligodendrocytes (PARP1), node of Ranvier and dorsal root ganglion (NCS1, FLRT3 and CDH15) and the innate immune system (CR1, AHSG and WARS) in multiple sclerosis. Our study demonstrates how harnessing large-scale genomic and proteomic data can yield new insights into the role of the plasma proteome in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lazaros Belbasis
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Sam Morris
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Derrick Bennett
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Robin Walters
- Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| |
Collapse
|
80
|
Factor SA, Weinshenker D, McKay JL. A possible pathway to freezing of gait in Parkinson's disease. JOURNAL OF PARKINSON'S DISEASE 2025; 15:282-290. [PMID: 39973500 DOI: 10.1177/1877718x241308487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Freezing of gait (FOG), a common, perplexing gait disorder observed in Parkinson's disease (PD), is a leading cause of injurious falls and contributes significantly to social isolation. Unlike other PD cardinal features, FOG appears to develop independently, and its heterogeneity presents challenges for both definition and measurement. The pathophysiological mechanisms underlying FOG remain poorly understood, limiting the development of effective treatments. Although the roles of specific, targetable biomarkers in FOG development remain unidentified, evidence suggests that it is likely multimodal, potentially involving extranigral transmitter circuits. The diversity of FOG phenotypes may also reflect underlying differences in pathophysiology. In this paper, we first present evidence that FOG may occur independently of dopaminergic influence. We then review an expanding body of research supporting the hypothesis that FOG arises from a dysfunctional pathophysiological feedback loop, involving norepinephrine (NE) depletion, neuroinflammation, and amyloid-β (Aβ) accumulation. This biological disruption occurs concurrently with, but distinct from, the primary dopaminergic pathology of PD. When they occur on the background of dopamine loss, the interactions between NE, Aβ, and inflammation, as observed in Alzheimer's disease models, may similarly play a critical role in the development of FOG in PD and could serve as pathobiological markers. The proposed changes in the pathophysiological loop might even precede its onset, highlighting the need for further investigation. A deeper understanding of the involvement of Aβ, NE, and inflammatory markers in FOG could pave the way for rapid clinical trials to test existing amyloid-clearing therapies and noradrenergic drugs in appropriate patient populations.
Collapse
Affiliation(s)
- Stewart A Factor
- Jean and Paul Amos Parkinson's Disease and Movement Disorder Program, Emory University, Atlanta, GA, USA
| | | | - J Lucas McKay
- Jean and Paul Amos Parkinson's Disease and Movement Disorder Program, Emory University, Atlanta, GA, USA
- Department of Biomedical Informatics, Emory University, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, GA, USA
| |
Collapse
|
81
|
Campomayor NB, Kim HJ, Kim M. Pro-Oxidative and Inflammatory Actions of Extracellular Hemoglobin and Heme: Molecular Events and Implications for Alzheimer's and Parkinson Disease. Biomol Ther (Seoul) 2025; 33:235-248. [PMID: 39962769 PMCID: PMC11893490 DOI: 10.4062/biomolther.2024.224] [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: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 03/01/2025] Open
Abstract
Hemoglobin (Hb) and heme, which are typically confined within red blood cells (RBCs), are essential for intravascular transport of gases and nutrients. However, these molecules acquire secondary functions upon exposure to the extracellular environment. Hb and heme generate reactive oxygen species (ROS), which are potent pro-inflammatory agents that contribute to oxidative stress and cellular damage. These events are relevant to neurodegenerative processes, where oxidative stress, irregular deposition of protein aggregates, and chronic inflammation are key pathological features. Extracellular Hb, heme, and oxidative stress derived from hemorrhagic events or RBC lysis may contribute to increased blood-brain barrier (BBB) permeability. These events allow Hb and heme to interact with neuroimmune cells and pathological protein aggregates, further amplifying pro-inflammatory signaling and the progression of Alzheimer's disease (AD) and Parkinson disease (PD). Chronic neuroinflammation, oxidative stress, and mitochondrial dysfunction lead to neuronal degeneration. Here, we sought to elucidate the pro-oxidative and inflammatory actions of extracellular Hb and heme, emphasizing their potential impact on AD and PD development.
Collapse
Affiliation(s)
- Nicole Bon Campomayor
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Chemistry & Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| | - Hee Jin Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Mikyung Kim
- Uimyung Research Institute for Neuroscience, Department of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Chemistry & Life Science, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|
82
|
Zare H, Kasdorf MM, Bakhshian Nik A. Microfluidics in neural extracellular vesicles characterization for early Alzheimer's disease diagnosis. Mol Cell Neurosci 2025; 132:103982. [PMID: 39631514 DOI: 10.1016/j.mcn.2024.103982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024] Open
Abstract
Dementia is a general term for conditions impairing cognitive abilities including perception, reasoning, attention, judgment, memory, and daily brain function. Early diagnosis of Alzheimer's disease (AD), the most common form of dementia, using neural extracellular vesicles (nEVs) is the focus of the current study. These nEVs carry AD biomarkers including β-amyloid proteins and phosphorylated tau proteins. The novelty of this review lies in developing a microfluidic perspective by introducing the techniques using a microfluidic platform for early diagnosis of AD. A microfluidic device can detect small sample sizes with significantly low concentrations. These devices combine nEV isolation, enrichment, and detection, which makes them ideal candidates for early AD diagnosis.
Collapse
Affiliation(s)
- Hossein Zare
- Chemical and Biochemical Engineering Department, The University of Iowa, Iowa City, IA 52242, USA.
| | | | | |
Collapse
|
83
|
Kim BS, Hwang I, Ko HR, Kim YK, Kim HJ, Seo SW, Choi Y, Lim S, Kim YK, Nie S, Ye K, Park JC, Lee Y, Jo DG, Lee SE, Kim D, Cho SW, Ahn JY. EBP1 potentiates amyloid β pathology by regulating γ-secretase. NATURE AGING 2025; 5:486-503. [PMID: 39779912 DOI: 10.1038/s43587-024-00790-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
The abnormal deposition of amyloid β (Aβ), produced by proteolytic cleavage events of amyloid precursor protein involving the protease γ-secretase and subsequent polymerization into amyloid plaques, plays a key role in the neuropathology of Alzheimer's disease (AD). Here we show that ErbB3 binding protein 1 (EBP1)/proliferation-associated 2G4 (PA2G4) interacts with presenilin, a catalytic subunit of γ-secretase, inhibiting Aβ production. Mice lacking forebrain Ebp1/Pa2g4 recapitulate the representative phenotypes of late-onset sporadic AD, displaying an age-dependent increase in Aβ deposition, amyloid plaques and cognitive dysfunction. In postmortem brains of patients with AD and 5x-FAD mice, we found that EBP1 is proteolytically cleaved by asparagine endopeptidase at N84 and N204 residues, compromising its inhibitory effect on γ-secretase, increasing Aβ aggregation and neurodegeneration. Accordingly, injection of AAV2-Ebp1 wild-type or an asparagine endopeptidase-uncleavable mutant into the brains of 5x-FAD mice decreased Aβ generation and alleviated the behavioral impairments. Thus, our study suggests that EBP1 acts as an inhibitor of γ-secretase on amyloid precursor protein cleavage and preservation of functional EBP1 could be a therapeutic strategy for AD.
Collapse
Affiliation(s)
- Byeong-Seong Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Inwoo Hwang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Hyo Rim Ko
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Young Kwan Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yujung Choi
- Center for Brain Disorders, Brain Science Institute Korea Institute of Science and Technology (KIST), Seoul, Korea
- Department of Life Sciences, Korea University, Seoul, Korea
| | - Sungsu Lim
- Center for Brain Disorders, Brain Science Institute Korea Institute of Science and Technology (KIST), Seoul, Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Yun Kyung Kim
- Center for Brain Disorders, Brain Science Institute Korea Institute of Science and Technology (KIST), Seoul, Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Korea
| | - Shuke Nie
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Keqiang Ye
- Faculty of Life and Health Sciences, and Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jong-Chan Park
- Department of Biophysics, Sungkyunkwan University, Suwon, Korea
| | - Yunjong Lee
- Department of Pharmacology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Seung Eun Lee
- Research Animal Resources Center, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Daesik Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Sung-Woo Cho
- Department of Biochemistry and Molecular Biology, University of Ulsan, College of Medicine, Seoul, Korea
| | - Jee-Yin Ahn
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea.
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
84
|
Fułek M, Hachiya N, Gachowska M, Beszłej JA, Bartoszewska E, Kurpas D, Kurpiński T, Adamska H, Poręba R, Urban S, Fułek K, Leszek J. Cellular Prion Protein and Amyloid-β Oligomers in Alzheimer's Disease-Are There Connections? Int J Mol Sci 2025; 26:2097. [PMID: 40076721 PMCID: PMC11900156 DOI: 10.3390/ijms26052097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. Pathological deposits of neurotoxin proteins within the brain, such as amyloid-β and hyperphosphorylated tau tangles, are prominent features in AD. The prion protein (PrP) is involved in neurodegeneration via its conversion from the normal cellular form (PrPC) to the infection prion protein scrapie (PrPSc) form. Some studies indicated that post-translationally modified PrPC isoforms play a fundamental role in AD pathological progression. Several studies have shown that the interaction of Aβ oligomers (Aβos) with the N-terminal residues of the PrPC protein region appears critical for neuronal toxicity. PrPC-Aβ binding always occurs in AD brains and is never detected in non-demented controls, and the binding of Aβ aggregates to PrPC is restricted to the N-terminus of PrPC. In this study, we aimed to gather all of the recent information about the connections between PrPC and AD, with potential clinical implications.
Collapse
Affiliation(s)
- Michał Fułek
- Department and Clinic of Diabetology, Hypertension and Internal Diseases, Institute of Internal Diseases, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Naomi Hachiya
- Shonan Research Center, New-STEP Research Center, Central Glass Co., Ltd., Shonan Health Innovation Park 26-1, Muraoka Higashi, Fujisawa 251-8555, Kanagawa, Japan;
| | - Martyna Gachowska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Jan Aleksander Beszłej
- Department and Clinic of Psychiatry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| | - Elżbieta Bartoszewska
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Donata Kurpas
- Division of Research Methodology, Department of Nursing, Faculty of Nursing and Midwifery, Wroclaw Medical University, 51-618 Wroclaw, Poland;
| | - Tomasz Kurpiński
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland; (M.G.); (E.B.); (T.K.)
| | - Hanna Adamska
- Department of Rheumatology and Internal Medicine, Marciniak Lower Silesian Specialist Hospital, 54-049 Wroclaw, Poland;
| | - Rafał Poręba
- Department of Biological Principles of Physical Activity, Wroclaw University of Health and Sport Sciences, 51-612 Wroclaw, Poland;
| | - Szymon Urban
- Department of Cardiology, The Copper Health Center, 59-301 Lubin, Poland;
| | - Katarzyna Fułek
- Department and Clinic of Otolaryngology, Head and Neck Surgery, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Jerzy Leszek
- Department and Clinic of Psychiatry, Wroclaw Medical University, 50-367 Wroclaw, Poland;
| |
Collapse
|
85
|
Shen M, Zhang C, Zhang Y, Lu D, Yuan J, Wang Z, Wu M, Zhu M, Chen Q. Mesoscale orchestration of collagen-based hierarchical mineralization. Nat Commun 2025; 16:2041. [PMID: 40016239 PMCID: PMC11868392 DOI: 10.1038/s41467-025-57189-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/14/2025] [Indexed: 03/01/2025] Open
Abstract
Mesoscale building blocks are instrumental in bridging multilevel hierarchical mineralization, endowing macroscale entities with remarkable functionality and mechanical properties. However, the mechanism orchestrating the homogeneous morphology of mesoscale mineralized motifs in collagen-based hard tissues remains unknown. Here, utilizing avian tendons as a mineralization model, we reveal a robust correlation between the mesoscale mineralized spherules and the presence of phosvitin. By designing a phosvitin-stabilized biomineral cluster medium, we replicate the well-defined mesoscale spherical structure within collagen matrix in vitro and ex vivo. In-depth studies reveal that phosvitin undergoes a conformational transition in the presence of biominerals at physiological concentrations, and self-assembles into mineral-dense amyloid-like aggregates. The spatial binding of these mineral-dense aggregates to collagen serves as a template for guiding the formation of mineralized spherules on the mesoscale. On the nanoscale, this binding facilitates mineral precursor release and diffusion into the fibrils for intrafibrillar mineralization. This discovery underscores the pivotal role of phosvitin-biomineral aggregates in templating hierarchical mineralization from the mesoscale to the nanoscale. This study not only elucidates the intricate mechanism underlying the collagen-based mineralization hierarchy but also promotes a cutting-edge advance in highly biomimetic material design and regenerative medicine.
Collapse
Affiliation(s)
- Minjuan Shen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Chunyan Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Yangyang Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Danyang Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Jian Yuan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Zhiyong Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Mengjie Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China
| | - Mengqi Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
86
|
Li CY, Fan LY, Lin CH, Hu CJ, Chiu MJ. Ultrasensitive Assays Detect Different Conformations of Plasma β Amyloids. ACS OMEGA 2025; 10:7256-7263. [PMID: 40028141 PMCID: PMC11865983 DOI: 10.1021/acsomega.4c10879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/18/2025] [Accepted: 01/28/2025] [Indexed: 03/05/2025]
Abstract
With the developments of ultrasensitive technologies such as immunomagnetic reduction (IMR) assay, single molecule array (SIMOA) assay, electrochemiluminescence immunoassay (ECLIA), the assay of blood-based amyloid 1-42 (Aβ1-42) becomes possible. However, the changes in measured plasma Aβ1-42 concentrations in Alzheimer's disease (AD) compared to cognitively unimpaired subjects (CU) are inconsistent. A possible reason for the inconsistency regarding various conformations of Aβ1-42 in plasma is explored in this study. Three samples with equal amounts of Aβ1-42 but different proportions of monomers and oligomers of Aβ1-42 were prepared. The Aβ1-42 composition of monomers and oligomers in samples was analyzed with Western blot. Identically diluted versions of these three samples were assayed with IMR and SIMOA for Aβ1-42 concentrations. The three diluted samples showed similar levels of Aβ1-42 assayed with IMR, whereas much lower levels for samples with more oligomers assayed with SIOMA. The results imply that IMR detects both monomers and oligomers of Aβ1-42. The measured levels of Aβ1-42 are independent of the proportions of monomer or oligomer Aβ1-42 but depend on the total amounts of Aβ1-42. In the case of SIMOA, monomers of Aβ1-42 are the primary target measured. By comparing Aβ1-42 concentrations of the plasma using IMR and SIMOA, the significant difference in plasma Aβ1-42 levels using IMR in AD compared to CU is mainly due to the formations of oligomeric Aβ1-42. Therefore, if the target molecules are monomers of Aβ1-42, SIMOA is the method of choice. Still, if the target molecules should include monomers, small and large oligomers, IMR would be an optimal consideration. In the future, the clinical implications of the proportion of oligomeric Aβ1-42 need to be elucidated.
Collapse
Affiliation(s)
- Chia-Yu Li
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ling-Yun Fan
- Departments
of Neurology, National Taiwan University
Hospital Bei-Hu Branch, Taipei 108, Taiwan
| | - Chin-Hsien Lin
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Institute
of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department
of Biomedical Engineering, National Taiwan
University, Taipei 106, Taiwan
| | - Chaur-Jong Hu
- Taipei
Neuroscience Institute, Taipei Medical University, New Taipei City, 235 Taiwan
- Department
of Neurology and Dementia Center, Taipei
Medical University-Shuang Ho Hospital, New Taipei City 235, Taiwan
- Department
of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Ming-Jang Chiu
- Department
of Neurology, National Taiwan University
Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
87
|
Pokrzyk J, Kulczyńska-Przybik A, Guzik-Makaruk E, Winkel I, Mroczko B. Clinical Importance of Amyloid Beta Implication in the Detection and Treatment of Alzheimer's Disease. Int J Mol Sci 2025; 26:1935. [PMID: 40076562 PMCID: PMC11900921 DOI: 10.3390/ijms26051935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
The role of amyloid beta peptide (Aβ) in memory regulation has been a subject of substantial interest and debate in neuroscience, because of both physiological and clinical issues. Understanding the dual nature of Aβ in memory regulation is crucial for developing effective treatments for Alzheimer's disease (AD). Moreover, accurate detection and quantification methods of Aβ isoforms have been tested for diagnostic purposes and therapeutic interventions. This review provides insight into the current knowledge about the methods of amyloid beta detection in vivo and in vitro by fluid tests and brain imaging methods (PET), which allow for preclinical recognition of the disease. Currently, the priority in the development of new therapies for Alzheimer's disease has been given to potential changes in the progression of the disease. In light of increasing amounts of data, this review was focused on the diagnostic and therapeutic employment of amyloid beta in Alzheimer's disease.
Collapse
Affiliation(s)
- Justyna Pokrzyk
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland; (J.P.); (B.M.)
| | - Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland; (J.P.); (B.M.)
| | | | - Izabela Winkel
- Dementia Disorders Centre, Medical University of Wroclaw, 50-425 Ścinawa, Poland;
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland; (J.P.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Białystok, 15-269 Bialystok, Poland
| |
Collapse
|
88
|
Atanasova M. Small-Molecule Inhibitors of Amyloid Beta: Insights from Molecular Dynamics-Part A: Endogenous Compounds and Repurposed Drugs. Pharmaceuticals (Basel) 2025; 18:306. [PMID: 40143085 PMCID: PMC11944459 DOI: 10.3390/ph18030306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The amyloid hypothesis is the predominant model of Alzheimer's disease (AD) pathogenesis, suggesting that amyloid beta (Aβ) peptide is the primary driver of neurotoxicity and a cascade of pathological events in the central nervous system. Aβ aggregation into oligomers and deposits triggers various processes, such as vascular damage, inflammation-induced astrocyte and microglia activation, disrupted neuronal ionic homeostasis, oxidative stress, abnormal kinase and phosphatase activity, tau phosphorylation, neurofibrillary tangle formation, cognitive dysfunction, synaptic loss, cell death, and, ultimately, dementia. Molecular dynamics (MD) is a powerful structure-based drug design (SBDD) approach that aids in understanding the properties, functions, and mechanisms of action or inhibition of biomolecules. As the only method capable of simulating atomic-level internal motions, MD provides unique insights that cannot be obtained through other techniques. Integrating experimental data with MD simulations allows for a more comprehensive understanding of biological processes and molecular interactions. This review summarizes and evaluates MD studies from the past decade on small molecules, including endogenous compounds and repurposed drugs, that inhibit amyloid beta. Furthermore, it outlines key considerations for future MD simulations of amyloid inhibitors, offering a potential framework for studies aimed at elucidating the mechanisms of amyloid beta inhibition by small molecules.
Collapse
|
89
|
Han X, Li PH, Wang S, Blakely T, Aggarwal S, Gopalani B, Sanchez M, Schalek R, Meirovitch Y, Lin Z, Berger D, Wu Y, Aly F, Bay S, Delatour B, Lafaye P, Pfister H, Wei D, Jain V, Ploegh H, Lichtman J. Mapping Alzheimer's Molecular Pathologies in Large-Scale Connectomics Data: A Publicly Accessible Correlative Microscopy Resource. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.24.563674. [PMID: 37961104 PMCID: PMC10634883 DOI: 10.1101/2023.10.24.563674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Connectomics using volume-electron-microscopy enables mapping and analysis of neuronal networks, revealing insights into neural circuit function and dysfunction. In Alzheimer's disease (AD), where amyloid-β (Aβ) and hyperphosphorylated-Tau (pTau) are implicated, connectomics offers an approach to unravel how these molecules contribute to circuit alterations by enabling the study of these molecules within the context of the complete local neuronal and glial milieu. We present a volumetric-correlated-light-and-electron microscopy (vCLEM) protocol using fluorescent nanobodies to localize Aβ and pTau within a large-scale connectomics dataset from the hippocampus of the 3xTg AD mouse model. A key outcome of this work is a publicly accessible vCLEM dataset, featuring fluorescent labeling of Aβ and pTau in the ultrastructural context with segmented neurons, glia, and synapses. This dataset provides a unique resource for exploring AD pathology in the context of connectomics and fosters collaborative opportunities in neurodegenerative disease research. As a proof-of-principle, we uncovered new localizations of Aβ and pTau, including pTau-positive spine-like protrusions at the axon initial segment and changes in the number and size of synapses near Aβ plaques. Our vCLEM approach facilitates the discovery of both molecular and structural alterations within large-scale EM data, advancing connectomics research in Alzheimer's and other neurodegenerative diseases.
Collapse
|
90
|
Gao Z, Hou R, Qian C. Functional Proteins/Peptides Targeting to Clear Amyloid-β for Alzheimer's Disease Therapy. Chembiochem 2025; 26:e202400912. [PMID: 39813167 DOI: 10.1002/cbic.202400912] [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: 11/01/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
Alzheimer's disease (AD) is a significant neurodegenerative disorder primarily affecting individuals over the age of 65. It is characterized by impairments in memory, thinking, analytical judgment, visuospatial recognition, and mood. In recent years, the development of protein and peptide drugs targeting amyloid-beta (Aβ) has gained momentum, with several therapies entering clinical trials and even receiving marketing approval. Novel functional protein and peptide drugs, as the first-generation immunotherapeutic agents for neurodegenerative diseases, have pioneered cellular immunotherapy for AD. However, the currently available drugs are associated with toxicity issues, which can lead to serious complications such as cerebral hemorrhage or edema. Consequently, this study examines the potential for a new generation of Aβ-targeting drugs to mitigate the side effects of existing treatments and offers innovative perspectives for the advancement of therapies for AD.
Collapse
Affiliation(s)
- Ziran Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Renjie Hou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| | - Chenggen Qian
- State Key Laboratory of Natural Medicines, Department of Pharmaceutical Science, School of Pharmacy, China Pharmaceutical University, Jiangsu, Nanjing, 210009, P.R. China
| |
Collapse
|
91
|
Medd MM, Yon JE, Dong H. RhoA/ROCK/GSK3β Signaling: A Keystone in Understanding Alzheimer's Disease. Curr Issues Mol Biol 2025; 47:124. [PMID: 39996845 PMCID: PMC11854763 DOI: 10.3390/cimb47020124] [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: 01/19/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive cognitive decline and loss of neuronal integrity. Emerging evidence suggests that RhoA, Rho-associated coiled-coil kinase (ROCK), and their downstream effector molecule glycogen synthase 3β (GSK3β) interact within a complex signaling pathway (RhoA/ROCK/GSK3β) that plays a crucial role in the pathogenesis of AD. RhoA, a small GTPase, along with its downstream effector, ROCK, regulates various cellular processes, including actin cytoskeleton dynamics, apoptosis, and synaptic plasticity. GSK3β, a serine/threonine kinase, plays a key role in neuronal function and AD pathology, including the regulation of tau phosphorylation and amyloid-beta cleavage. Overactive GSK3β has been closely linked to tau hyperphosphorylation, neurodegeneration, and the progression of AD. Thus, GSK3β has been considered as a promising therapeutic target for treating AD and mitigating cognitive impairment. However, clinical trials of GSK3β in AD have faced considerable challenges due to the complexity of the specific neuronal inhibition of GSK3β. In this review, we summarize the literature regarding the relationship of RhoA/ROCK and GSK3β signaling pathways in AD pathogenesis. We further discuss recent findings of the sTREM2-transgelin-2 (TG2) axis as a potential mediator of this complex pathway and provide our review on a novel targeting strategy for AD.
Collapse
Affiliation(s)
- Milan M. Medd
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.M.M.); (J.E.Y.)
| | - Jayden E. Yon
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.M.M.); (J.E.Y.)
| | - Hongxin Dong
- Stephen M. Stahl Center for Psychiatric Neuroscience, Departments of Psychiatry & Behavioral Sciences and Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
92
|
Bergendorf A, Park JH, Ball BK, Brubaker DK. Mouse-to-human modeling of microglia single-nuclei transcriptomics identifies immune signaling pathways and potential therapeutic candidates associated with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637100. [PMID: 39975195 PMCID: PMC11839086 DOI: 10.1101/2025.02.07.637100] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and behavior change. Studies have found that dysregulation of microglial cells is pivotal to AD pathology. These mechanisms have been studied in mouse models to uncover potential therapeutic biomarkers. Despite these findings, there are limitations to the translatable biological information from mice to humans due to differences in physiology, timeline of disease, and the heterogeneity of humans. To address the inter-species discrepancies, we developed a novel implementation of the Translatable Components Regression (TransComp-R) framework, which integrated microglia single-nuclei mouse and human transcriptomics data to identify biological pathways in mice predictive of human AD. We compared model variations with sparse and traditional principal component analysis. We found that both dimensionality reduction techniques encoded similar AD disease biology on mouse principal components with limited differences in technical performance. Several mouse sparse principal components explained high amounts of variance in humans and significantly differentiated human AD from control microglial cells. Additionally, we identified FDA-approved medications that induced gene expression profiles correlated with projections of healthy human microglia on mouse principal components. Such medications included cabergoline, selumetinib, and palbociclib. This computational framework may support uncovering cross-species disease insights and candidate pharmacological solutions from single-cell datasets.
Collapse
Affiliation(s)
- Alexander Bergendorf
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jee Hyun Park
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Brendan K. Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Douglas K. Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- The Blood, Heart, Lung, and Immunology Research Center, Case Western Reserve University, University Hospitals of Cleveland, Cleveland, OH 44106, USA
| |
Collapse
|
93
|
Tiwari A, Singh R, Kumar S, Sunkaria A, Jain A. From Plant to Pathway: Molecular Mechanisms of Ruscogenin in Preventing Amyloid-Beta Aggregation through Computational and Experimental Approaches. ACS Chem Neurosci 2025; 16:500-512. [PMID: 39793029 DOI: 10.1021/acschemneuro.4c00745] [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: 01/12/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, extracellular amyloid-β (Aβ) plaque accumulation, and intracellular neurofibrillary tangles. Recent efforts to find effective therapies have increased interest in natural compounds with multifaceted effects on AD pathology. This study explores natural compounds for their potential to mitigate AD pathology using molecular docking, ADME screening, and in vitro assays, with ruscogenin─a steroidal sapogenin from Ruscus aculeatus─emerging as a promising candidate. Ruscogenin, known for its antioxidant and anti-inflammatory properties, was investigated for its effects on Aβ aggregation, a critical process in AD progression. In vitro assays demonstrated that ruscogenin inhibits Aβ oligomerization at equimolar and higher molar ratios. Molecular dynamics (MD) simulations further revealed that ruscogenin targets aggregation-prone regions, reducing noncovalent interactions and the solvent-accessible surface area of Aβ aggregates. These effects were concentration-dependent, with higher concentrations yielding optimal inhibition, pointing to a multiphasic behavior in ruscogenin's modulation of Aβ aggregation. This study highlights ruscogenin's potential as a natural therapeutic agent for AD, capable of addressing both oxidative stress and inflammation. The findings lay the groundwork for further exploration of ruscogenin-based interventions and underscore the broader potential of natural compounds in AD treatment strategies.
Collapse
Affiliation(s)
- Aastha Tiwari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology Mesra, Ranchi, Jharkhand 835215, India
| | - Ravinder Singh
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Shubham Kumar
- Department of Bioengineering and Biotechnology, Birla Institute of Technology Mesra, Ranchi, Jharkhand 835215, India
| | - Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Alok Jain
- Department of Bioengineering and Biotechnology, Birla Institute of Technology Mesra, Ranchi, Jharkhand 835215, India
| |
Collapse
|
94
|
Ogunro OB, Karigidi ME, Gyebi GA, Turkistani A, Almehmadi AH. Tangeretin offers neuroprotection against colchicine-induced memory impairment in Wistar rats by modulating the antioxidant milieu, inflammatory mediators and oxidative stress in the brain tissue. BMC Complement Med Ther 2025; 25:40. [PMID: 39905390 PMCID: PMC11792585 DOI: 10.1186/s12906-025-04769-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 01/17/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Tangeretin, a flavone compound (O-polymethoxylated) naturally present in tangerine and other citrus peels has demonstrated effectiveness as an anti-inflammatory and neuroprotective agent in several disease model. This study evaluated the impact of tangeretin in mitigating cognitive dysfunction and oxidative stress induced by colchicine in rats, comparing its efficacy with donepezil hydrochloride. METHODS Cognitive dysfunction was induced by administering colchicine (15 µg/rat) intracerebroventricularly (ICV) via a stereotaxic apparatus in male Wistar rats. Colchicine resulted in poor memory retention in acquiring and retaining a spatial navigation task, passive avoidance apparatus, and Morris water maze paradigms. Chronic treatment with tangeretin (at doses of 50, 100, and 200 mg/kg, p.o. once daily) and donepezil hydrochloride (at a dose of 10 mg/kg, p.o. daily) for 28 days, starting seven days before colchicine injection, significantly ameliorated colchicine-induced cognitive impairment. RESULTS The biochemical analysis showed that chronic administration of tangeretin effectively reversed the colchicine-induced increase in the level/activity of lipid peroxidation, hydrogen peroxide (H2O2), myeloperoxidase (MPO), nitrite, reactive oxygen species (ROS), tumour necrosis factor-α (TNF-α), nuclear factor kappa B (NF-κB), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-10 (IL-10), serotonin, dopamine, glutamate, amyloid beta (Aβ) peptide, and caspase-3. Tangeretin also reversed the colchicine-induced reduction in the level/activity of brain-derived neurotrophic factor (BDNF), amma-aminobutyric acid (GABA), acetylcholinesterase (AChE), glutathione S-Transferase (GST), glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT), superoxide dismutase (SOD), reduced glutathione (GSH), and total thiol (T-SH) in rat brains. However, donepezil hydrochloride did not prevent oxidative stress. CONCLUSIONS These findings suggest that chronic administration of tangeretin at 50, 100, and 200 mg/kg, p.o. once daily, was protective in mitigating colchicine-induced cognitive impairment and associated oxidative stress. At the same time, donepezil hydrochloride did not demonstrate similar effects.
Collapse
Affiliation(s)
- Olalekan Bukunmi Ogunro
- Pharmacology, Drug Discovery and Toxicology Research Laboratory, Department of Biological Sciences, KolaDaisi University, Ibadan, 200213, Nigeria.
| | - Mojisola Esther Karigidi
- Pharmacology, Drug Discovery and Toxicology Research Laboratory, Department of Biological Sciences, KolaDaisi University, Ibadan, 200213, Nigeria
| | - Gideon Ampoma Gyebi
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, P.O. Box 1334, Durban, 4000, South Africa
| | - Areej Turkistani
- Department of Pharmacology and Toxicology, College of Medicine, Taif University, Taif, 21944, Kingdom of Saudi Arabia
| | - Ahmad H Almehmadi
- Oral Biology Department, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
95
|
Gao T, Yan N, Pu Y, Zhang Z, Duan Z, Tang Z, Huang D, Chen Y, Yuan S, Yan X, Yuan M. Ginger leaf polyphenols mitigate β-amyloid toxicity via JNK/FOXO pathway activation in Caenorhabditis elegans. Food Funct 2025; 16:1072-1085. [PMID: 39829385 DOI: 10.1039/d4fo03238a] [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
β-Amyloid (Aβ) aggregation is the major pathological feature of Alzheimer's disease (AD), resulting in oxidative stress and further exacerbating Aβ aggregation. Ginger leaf polyphenols (GLP) have been found to possess antioxidant activity, evidencing their potential in addressing AD. GLP is mainly composed of 12 polyphenols, 8 organic acids, and 6 glycosides, of which polyphenols are predominantly composed of apigenin, kaempferol, and quercetin derivatives. Moreover, GLP alleviates reproductive toxicity, longevity toxicity, and neurotoxicity induced by Aβ via regulating the antioxidase system in Caenorhabditis elegans. As shown by the network pharmacology results, GLP might activate the JNK/Foxo signaling pathway to regulate the antioxidase system, which was evidenced by the up-regulation of gene expression levels of jnk-1, daf-16, sod-3, and hsp-16.2. Overall, GLP might be a potential antioxidant for combating AD.
Collapse
Affiliation(s)
- Tao Gao
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Ningning Yan
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Yaying Pu
- Yaan People's Hospital, Yaan, 625099, China.
| | - Zhonghao Zhang
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Zhihao Duan
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Zizhong Tang
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Daojian Huang
- Dazhu County Scientific and Technical Information Institute, Dazhou, 635100, China
| | - Yanger Chen
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| | - Shu Yuan
- College of Resources, Sichuan Agricultural University, Chengdu, 611130, China
| | | | - Ming Yuan
- College of Life Science, Sichuan Agricultural University, Yaan 625014, China.
| |
Collapse
|
96
|
Bao L, Li X, Tian J, Wang L, Ji Y, Cui Y, Sun W, Zhang J, Xia M, Zhu P, Cui G, Chen H. GGC repeat expansions in NOTCH2NLC cause uN2CpolyG cerebral amyloid angiopathy. Brain 2025; 148:467-479. [PMID: 39167540 DOI: 10.1093/brain/awae274] [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: 04/15/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
The expansion of GGC repeats within NOTCH2NLC leads to translation of the uN2CpolyG protein, the primary pathogenic factor in neuronal intranuclear inclusion disease (NIID). The aim of this study was to explore the deposition of uN2CpolyG as an amyloid in the vessel wall, leading to uN2CpolyG cerebral amyloid angiopathy-related cerebral microbleeds (CMBs). A total of 97 patients with genetically confirmed NIID were enrolled in this study. We analysed the presence of CMBs using susceptibility-weighted imaging sequences and compared general clinical information, cerebrovascular risk factors, stroke history, antiplatelet medication use and MRI features between NIID patients with and without CMBs. We also performed haematoxylin and eosin, Perl's, Congo red and Thioflavin S staining, ubiquitin, p62 and uN2CpolyG immunostaining on brain tissue obtained from four NIID patients. A total of 354 CMBs were detected among 41 patients with NIID, with nearly half located in the deep brain, one-third in the lobes and ∼20% in the infratentorial area. No significant differences in cerebrovascular disease risk factors or history of antiplatelet drug use were observed between patients with and without CMBs. However, patients with CMBs had suffered a higher incidence of previous ischaemic and haemorrhagic stroke events. This group also had a higher incidence of recent subcortical infarcts and a higher proportion of white matter lesions in the external capsule and temporal pole. Conversely, patients without CMBs showed higher detection of high signals at the corticomedullary junction on diffusion-weighted imaging and more pronounced brain atrophy. Haematoxylin and eosin staining showed blood vessel leakage and haemosiderin-laden macrophage clusters, and Prussian blue staining revealed iron deposition in brain tissue. CMBs occurred more frequently in small vessels lacking intranuclear inclusions, and extensive degeneration of endothelial cells and smooth muscle fibres was observed mainly in vessels lacking inclusions. Congo red-positive amyloid deposition was observed in the cerebral vessels of NIID patients, with disordered filamentous fibres appearing under an electron microscope. Additionally, the co-localization of Thioflavin S-labelled amyloid and uN2CpolyG protein in the cerebral vascular walls of NIID patients further suggested that uN2CpolyG is the main pathogenic protein in this form of amyloid angiopathy. In conclusion, we reviewed patients with GGC repeat expansion of NOTCH2NLC from a new perspective, providing initial clinical, neuroimaging and pathological evidence suggesting that uN2CpolyG might contribute to a distinct type of cerebral amyloid angiopathy.
Collapse
Affiliation(s)
- Lei Bao
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiaowen Li
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Jin Tian
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Lulu Wang
- Department of Pathology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Ying Ji
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Yingying Cui
- Department of Pathology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Wen Sun
- Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Jing Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100000, China
| | - Man Xia
- Department of Neurological Intensive Care, Yantai Yuhuangding Hospital, Yantai 264000, China
| | - Pinyi Zhu
- Department of Geriatric Neurology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing 210000, China
| | - Guiyun Cui
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
- Department of Neurology, Xuzhou Medical University, Xuzhou 221004, China
| | - Hao Chen
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
97
|
AmeliMojarad M, AmeliMojarad M, Cui X. An overview on the impact of viral pathogens on Alzheimer's disease. Ageing Res Rev 2025; 104:102615. [PMID: 39631533 DOI: 10.1016/j.arr.2024.102615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/30/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia which affects over than 60 million cases worldwide with higher incidence in low and middle-income countries by 2030. Based on the multifactorial nature of AD different risk factors are linked to the condition considering the brain's β-amyloid plaques (Aβ) and neurofibrillary tangles (NFTs) as its primary hallmarks. Lately, viral photogenes specially after recent SARS-CoV-2 pandemic has gained a lot of attention in promoting the neurodegenerative disorder such as AD based on their capacity to increase the permeability of the blood-brain barrier, dysregulation of immune responses, and the impact on Aβ processing and phosphorylation of tau proteins. Therefore, in this review, we summarized the important association of viral pathogens and their mechanism by which they contribute with AD formation and development. AN OVERVIEW OF THE ROLES OF VIRAL PATHOGENS IN AD: According to this figure, viruses can infect neurons directly by modulating the BBB, transferring from endothelial cells to glial cells and then to neurons, increasing the Aβ deposition, and affecting the tau protein phosphorylation or indirectly through the virus's entrance and pathogenicity that can be accelerated by genetic and epigenetic factors, as well as chronic neuroinflammation caused by activated microglia and astrocytes.
Collapse
Affiliation(s)
- Melika AmeliMojarad
- Department of Oncology, The First Affiliate Hospital of Dalian Medical University, Zhongshan Road, Dalian, Liaoning 116011, China.
| | - Mandana AmeliMojarad
- Department of Oncology, The First Affiliate Hospital of Dalian Medical University, Zhongshan Road, Dalian, Liaoning 116011, China.
| | - Xiaonan Cui
- Department of Oncology, The First Affiliate Hospital of Dalian Medical University, Zhongshan Road, Dalian, Liaoning 116011, China.
| |
Collapse
|
98
|
Anitha K, Singh MK, Kohat K, Sri Varshini T, Chenchula S, Padmavathi R, Amerneni LS, Vishnu Vardhan K, Mythili Bai K, Chavan MR, Bhatt S. Recent Insights into the Neurobiology of Alzheimer's Disease and Advanced Treatment Strategies. Mol Neurobiol 2025; 62:2314-2332. [PMID: 39102108 DOI: 10.1007/s12035-024-04384-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024]
Abstract
In recent years, significant advancements have been made in understanding Alzheimer's disease from both neurobiological and clinical perspectives. Exploring the complex systems underlying AD has unveiled insights that could potentially revolutionize therapeutic approaches. Recent investigations have highlighted intricate interactions among genetic, molecular, and environmental factors in AD. Optimism arises from neurobiological advancements and diverse treatment options, potentially slowing or halting disease progression. Amyloid-beta plaques and tau protein tangles crucially influence AD onset and progression. Emerging treatments involve diverse strategies, such as approaches targeting multiple pathways involved in AD pathogenesis, such as inflammation, oxidative stress, and synaptic dysfunction pathways. Clinical trials using humanized monoclonal antibodies, focusing on immunotherapies eliminating amyloid-beta, have shown promise. Nonpharmacological interventions such as light therapy, electrical stimulation, cognitive training, physical activity, and dietary changes have drawn attention for their potential to slow cognitive aging and enhance brain health. Precision medicine, which involves tailoring therapies to individual genetic and molecular profiles, has gained traction. Ongoing research and interdisciplinary collaboration are expected to yield more effective treatments.
Collapse
Affiliation(s)
- Anitha K
- School of Pharmacy and Technology Management (SPTM), SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed to University, Shirpur, 425405, India
| | | | - Komal Kohat
- All India Institute of Medical Sciences, Madhya Pradesh, Bhopal, 462020, India
| | - Sri Varshini T
- All India Institute of Medical Sciences, Raipur, 462020, India
| | - Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, 462020, India.
| | - Padmavathi R
- SVS Medical College, Hyderabad, Telangana, India
| | | | - Vishnu Vardhan K
- All India Institute of Medical Sciences, Madhya Pradesh, Bhopal, 462020, India
| | | | - Madhav Rao Chavan
- All India Institute of Medical Sciences, Mangalagiri, Andhra Pradesh, India
| | - Shvetank Bhatt
- School of Health Sciences and Technology, MIT World Peace University, Dr. Vishwanath Karad, Pune, 411038, Maharashtra, India
| |
Collapse
|
99
|
Titisari N, Ahmad H, Samsulrizal N, Fauzi A, Abdul Razak IS. The mechanism underlying streptozotocin injection for the development of a nontransgenic Alzheimer's disease animal model. Open Vet J 2025; 15:594-600. [PMID: 40201829 PMCID: PMC11974322 DOI: 10.5455/ovj.2025.v15.i2.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/02/2025] [Indexed: 04/10/2025] Open
Abstract
Streptozotocin (STZ) is a widely used chemical agent in biomedical research. It is primarily known for its ability to induce high blood glucose levels in animal models by selectively destroying pancreatic beta cells. Nonetheless, many studies have also used STZ to generate animal models of diabetic complications, such as Alzheimer's disease (AD) animal models. STZ induction promotes hyperglycemia, which activates numerous mechanism pathways that result in the production of pathogenic AD characteristics, including beta-amyloid accumulation and neurofibrillary tangles. Numerous theories exist to elucidate the mechanisms underlying diabetes and AD; however, studies on the potential of an animal model of STZ-induced AD remain limited. Thus, this review summarizes the pathogenesis associated with STZ exposure, particularly in AD animal model studies related to diabetes. More specifically, this study will discuss the relationship between increased blood glucose levels after STZ injection and the process of beta-amyloid formation and insulin dysfunction in the brain.
Collapse
Affiliation(s)
- Nurina Titisari
- Department of Veterinary Physiology, Faculty of Veterinary Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Hafandi Ahmad
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang,Malaysia
| | | | - Ahmad Fauzi
- Department of Veterinary Clinical Pathology, Faculty of Veterinary Medicine, Universitas Brawijaya,Malang, Indonesia
| | - Intan Shameha Abdul Razak
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang,Malaysia
| |
Collapse
|
100
|
Ajaikumar A, Watanabe NM, Suga K, Okamoto Y, Umakoshi H. Amyloid beta (Aβ) fibrillation kinetics and its impact on membrane polarity. J Bioenerg Biomembr 2025; 57:1-10. [PMID: 39760991 DOI: 10.1007/s10863-024-10046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025]
Abstract
Fibrillation of the amyloid beta (Aβ) peptide has often been associated with neurodegenerative pathologies such as Alzheimer's disease. In this study we examined the influence of several potential compositions of the lipid membrane on Aβ fibrillation by using liposomes as a basic model membrane. Firstly, it was revealed that Aβ fibrillation kinetics were enhanced and had the potential to occur at a faster rate on more fluid membranes compared to solid membranes. Next, the extent of fibril-related damage to membranes was examined with analysis of membrane polarity via the steady-state emission spectra of 6-dodecanoyl-2-dimethylaminonaphthalene (Laurdan). It was revealed that there was slight hydration behavior of the membrane during the lag phase (tlag) of the kinetic process, possibly coinciding with Aβ monomer binding. However, as the fibrillation kinetic process continued the membrane gradually dehydrated. Hydration states of membranes during and after Aβ fibrillation processes were further examined via deconvolution analysis of the obtained Laurdan spectra. This allows a mapping of membrane hydration from the interior to exterior regions of the lipid membrane. Results revealed slight but definitive variations in deeper region membrane polarity during the time course of Aβ fibrillation, suggesting Aβ aggregation impacts not only the surface level aggregating region but also the inner regions of the membrane. These results can ultimately contribute to the future investigations of the nature of the membrane damage caused by Aβ aggregation.
Collapse
Affiliation(s)
- Arun Ajaikumar
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka, 560-8531, Japan
| | - Nozomi Morishita Watanabe
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka, 560-8531, Japan
| | - Keishi Suga
- Department of Chemical Engineering, Tohoku University, Sendai, Miyagi, 980-8579, Japan
| | - Yukihiro Okamoto
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka, 560-8531, Japan
| | - Hiroshi Umakoshi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka, 560-8531, Japan.
| |
Collapse
|