1
|
Ma Y, Wang G, Chen H, Tsai M. Exploring Abeta42 monomer diffusion dynamics on fibril surfaces through molecular simulations. Protein Sci 2025; 34:e70131. [PMID: 40371804 PMCID: PMC12079388 DOI: 10.1002/pro.70131] [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: 11/22/2024] [Revised: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025]
Abstract
This study provides critical insights into the role of surface-mediated processes in Alzheimer's disease, with implications for the aggregation of Abeta42 peptides. Employing coarse-grained molecular dynamics simulations, we focus on elucidating the molecular intricacies of these processes beyond primary nucleation. Central to our investigation is the analysis of a freely diffusing Abeta42 monomer on preformed fibril structures. We conduct detailed calculations of the monomer's diffusion coefficient on fibril surfaces (as a one-dimensional case), along with various monomer orientations. Our findings reveal a strong and consistent correlation between the monomer's diffusion coefficient and its orientation on the surface. Further analysis differentiates the effects of parallel and perpendicular alignments with respect to the fibril axis. Additionally, we explore how different fibril surfaces influence monomer dynamics by comparing the C-terminal and N-terminal surfaces. We find that the monomer exhibits faster diffusion coefficients on the C-terminal surface. Differences in surface roughness (SR), quantified using root-mean-square distances, significantly affect monomer dynamics, thereby influencing its diffusion on the surface. Importantly, this study underscores that fibril twisting acts as a regulatory niche, selectively influencing these orientations and their diffusion properties necessary for facilitating fibril growth within biologically relevant time scales. This discovery opens new avenues for targeted therapeutic strategies aimed at manipulating fibril dynamics to mitigate the progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Yuan‐Wei Ma
- Institute of Bioinformatics and Structural BiologyNational Tsing‐Hua UniversityHsinchuTaiwan
- Department of Chemistry and BiochemistryNational Chung Cheng UniversityChiayiTaiwan
| | - Guan‐Fang Wang
- Department of Chemistry and BiochemistryNational Chung Cheng UniversityChiayiTaiwan
| | - Hong‐Yi Chen
- Department of Chemistry and BiochemistryNational Chung Cheng UniversityChiayiTaiwan
| | - Min‐Yeh Tsai
- Department of Chemistry and BiochemistryNational Chung Cheng UniversityChiayiTaiwan
- Center for Nano Bio‐DetectionNational Chung Cheng UniversityChiayiTaiwan
- Division of Physics, National Center for Theoretical SciencesNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
2
|
Latina V, De Introna M, Malerba F, Florio R, Balzamino BO, Di Natale G, Sciacca MFM, Pappalardo G, Micera A, Pignataro A, Calissano P, Amadoro G. Acute targeting of N-terminal tau protein has long-lasting beneficial effects in Tg2576 APP/Aβ mouse model by reducing cognitive impairment, cerebral Aβ-amyloidosis, synaptic remodeling and microgliosis later in life. Acta Neuropathol Commun 2025; 13:121. [PMID: 40442822 PMCID: PMC12123992 DOI: 10.1186/s40478-025-02022-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: 03/13/2025] [Accepted: 04/30/2025] [Indexed: 06/02/2025] Open
Abstract
Even though the number of patients suffering from Alzheimer's Disease (AD) is rapidly growing worldwide, only a few symptomatic treatments have been approved for clinical use, pointing out the urgent need for more effective disease-modifying therapies that actually alter the progression of this neurodegenerative disorder which is characterized by co-occurence of both Amyloid beta (Aβ) and tau neuropathologies. Preclinical and clinical evidence suggests that a link between Aβ and tau drives the entire continuum of AD pathobiology. 12A12 is a monoclonal antibody (mAb) which offers neuroprotection into two transgenic lines of AD, including Tg2576 that overexpresses Swedish mutation (KM670/671NL) of Amyloid Precursor Protein (APP, isoform 695) and 3xTg (APP Swedish, MAPT P301L, and PSEN1 M146V), by targeting the 20-22kDa N-terminal tau fragments (NH2htau). In particular, acute (over 14 days with 4 doses), intravenous injection of 12A12mAb leads to significant improvement of cognitive, biochemical and histopathological AD signs in symptomatic 6-month-old Tg2576, a well-established transgenic mouse model that mimics the human amyloidosis with an age-dependent Aβ accumulation/aggregation and plaque deposition. Here, we report that Tg2576 mice, immunized with 12A12mAb at 6 months of age and returned to their home cage for additional 3 months, exhibit preserved spatial memory despite the anticipated interruption of antibody administration (discontinuous treatment). This enduring beneficial effect on memory deficit (up to 90 days after the last injection) is accompanied by normalization in the synaptic imbalance and microgliosis along with decrease of the most toxic A11-positive prefibrillar oligomers and inverse increase in 4kDa monomeric form(s) of Aβ 1-42. These findings reveal that: (i) soluble, pathogenetic tau specie(s) located at the N-terminal domain of protein early synergizes with Aβ in driving the progression of AD neuropathology; (ii) transient immunoneutralization of the NH2htau following short-term treatment with 12A12mAb exerts preventive, long-lasting neuroprotective effects, at least in part by interfering at "pre-plaque" stage with the progressive deposition of insoluble, fibrillar Aβ via a shift of its aggregation pathway into its less harmful, unaggregated monomeric forms. Taken together, these findings represent a strong rationale for the advancement of 12A12mAb to clinical stage aiming at preventing the Aβ-dependent neurodegeneration by lowering the cerebral levels of NH2htau in humans suffering from chronic, slow-progressing AD.
Collapse
Affiliation(s)
- Valentina Latina
- Institute of Translational Pharmacology (IFT)-National Research Council (CNR), Via Fosso del Cavaliere 100, 00133, Rome, Italy
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy
| | - Margherita De Introna
- Centro Di Ricerca Europeo Sul Cervello (CERC), IRCCS Santa Lucia Foundation (FSL), Via Fosso del Fiorano 43-44, 00143, Rome, Italy
- Department of Systems Medicine, University of Tor Vergata, Via Montpellier, 1, 00133, Rome, Italy
| | - Francesca Malerba
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy
- Institute of Nanotechnology Campus Ecotekne- National Research Council (CNR), Via Monteroni, 73100, Lecce, Italy
| | - Rita Florio
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy
| | - Bijorn Omar Balzamino
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184, Rome, Italy
| | - Giuseppe Di Natale
- Institute of Crystallography (IC)-National Research Council (CNR), Via Paolo Gaifami 18, 95126, Catania, Italy
| | | | - Giuseppe Pappalardo
- Institute of Crystallography (IC)-National Research Council (CNR), Via Paolo Gaifami 18, 95126, Catania, Italy
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo, 6, 00184, Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology (IFT)-National Research Council (CNR), Via Fosso del Cavaliere 100, 00133, Rome, Italy
- Centro Di Ricerca Europeo Sul Cervello (CERC), IRCCS Santa Lucia Foundation (FSL), Via Fosso del Fiorano 43-44, 00143, Rome, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy
| | - Giuseppina Amadoro
- Institute of Translational Pharmacology (IFT)-National Research Council (CNR), Via Fosso del Cavaliere 100, 00133, Rome, Italy.
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161, Rome, Italy.
| |
Collapse
|
3
|
Nong Y, Kim JS, Jia L, Arancio O, Wang Q. The interaction between neurotransmitter receptor activity and amyloid-β pathology in Alzheimer's disease. J Alzheimers Dis 2025:13872877251342273. [PMID: 40388923 DOI: 10.1177/13872877251342273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
The accumulation of amyloid-β (Aβ) peptides is a hallmark of Alzheimer's disease (AD). Central to AD pathology is the production of Aβ peptides through the amyloidogenic processing of amyloid-β protein precursor (AβPP) by β-secretase (BACE-1) and γ-secretase. Recent studies have shifted focus from Aβ plaque deposits to the more toxic soluble Aβ oligomers. One significant way in which Aβ peptides impair neuronal information processing is by influencing neurotransmitter receptor function. These receptors, including adrenergic, acetylcholine, dopamine, 5-HT, glutamate, and gamma-aminobutyric acid (GABA) receptors, play a crucial role in regulating synaptic transmission, which underlies perceptual and cognitive functions. This review explores how Aβ interacts with these key neurotransmitter receptors and how these interactions contribute to neural dysfunction in AD. Moreover, we examine how agonists and antagonists of these receptors influence Aβ pathology, offering new perspectives on potential therapeutic strategies to curb AD progression effectively and improve patients' quality of life.
Collapse
Affiliation(s)
- Yuhan Nong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jung Soo Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Litian Jia
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Ottavio Arancio
- Departments of Pathology & Cell Biology, and Medicine, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Qi Wang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Neurosurgery, Columbia University, New York, NY, USA
| |
Collapse
|
4
|
Godoy SR, Sanchis P, Frau J, Vilanova B, Adrover M. On the Potential Role of Phytate Against Neurodegeneration: It Protects Against Fe 3+-Catalyzed Degradation of Dopamine and Ascorbate and Against Fe 3+-Induced Protein Aggregation. Int J Mol Sci 2025; 26:4799. [PMID: 40429940 PMCID: PMC12112605 DOI: 10.3390/ijms26104799] [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: 03/25/2025] [Revised: 04/30/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Myo-inositol-1,2,3,4,5,6-hexakisphosphate (IP6) is commonly found in plant-derived foods and has important pharmacological properties against many pathologies. One of them appears to be neurodegeneration, which is notably stimulated by dysregulated metal metabolism. Consequently, we explore the role of IP6 in mitigating neurodegenerative events catalyzed by dysregulated free iron. More precisely, we performed spectrophotometric measurements in aqueous solutions to investigate the ability of IP6 to chelate Fe3+ and inhibit its role in catalyzing the oxidative degradation of dopamine and ascorbic acid, two key molecules in neuronal redox systems. Our results demonstrate that IP6 effectively prevents the formation of harmful intermediates, such as neuromelanin and reactive oxygen species, which are linked to neuronal damage. Additionally, we assessed the effect of IP6 on Fe3+-induced protein aggregation, focusing on α-synuclein, which is closely associated with Parkinson's disease. Our data reveal that IP6 accelerates the conversion of toxic α-synuclein oligomers into less harmful amyloid fibrils, thereby reducing their neurotoxic potential. Our findings highlight the dual function of IP6 as a potent Fe3+ chelator and modulator of protein aggregation pathways, reinforcing its potential as a neuroprotective agent. Consequently, IP6 offers promising therapeutic potential for mitigating the progression of neurodegenerative disorders such as Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Samantha Rebeca Godoy
- Interdisciplinary Group on Neurodegeneration, Vascular and Metabolic Diseases (INNoVAM), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain; (S.R.G.); (P.S.); (J.F.); (B.V.)
- Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa 79, E-07010 Palma de Mallorca, Spain
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Pilar Sanchis
- Interdisciplinary Group on Neurodegeneration, Vascular and Metabolic Diseases (INNoVAM), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain; (S.R.G.); (P.S.); (J.F.); (B.V.)
- Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa 79, E-07010 Palma de Mallorca, Spain
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan Frau
- Interdisciplinary Group on Neurodegeneration, Vascular and Metabolic Diseases (INNoVAM), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain; (S.R.G.); (P.S.); (J.F.); (B.V.)
- Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa 79, E-07010 Palma de Mallorca, Spain
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Bartolomé Vilanova
- Interdisciplinary Group on Neurodegeneration, Vascular and Metabolic Diseases (INNoVAM), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain; (S.R.G.); (P.S.); (J.F.); (B.V.)
- Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa 79, E-07010 Palma de Mallorca, Spain
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| | - Miquel Adrover
- Interdisciplinary Group on Neurodegeneration, Vascular and Metabolic Diseases (INNoVAM), Departament de Química, Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain; (S.R.G.); (P.S.); (J.F.); (B.V.)
- Health Research Institute of the Balearic Islands (IdISBa), Ctra. Valldemossa 79, E-07010 Palma de Mallorca, Spain
- Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, Ctra. Valldemossa km 7.5, E-07122 Palma de Mallorca, Spain
| |
Collapse
|
5
|
Oldam J, Tchernyshyov I, Van Eyk J, Troncoso J, Glabe CG, Agnetti G. Thioflavin T in-gel staining for ex vivo analysis of cardiac amyloid. Front Mol Biosci 2025; 12:1505250. [PMID: 40433590 PMCID: PMC12106040 DOI: 10.3389/fmolb.2025.1505250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/14/2025] [Indexed: 05/29/2025] Open
Abstract
There are limited options to quantify and characterize amyloid species from biological samples in a simple manner. Thioflavin T (ThT) has been used for decades to stain amyloid fibrils, but to our knowledge, we were the first to use it in-gel. Thioflavin T in-gel staining is convenient as it is fast, inexpensive, accessible to most laboratories, and compatible with other fluorescent stains and downstream analyses such as mass spectrometry (MS).
Collapse
Affiliation(s)
- Joseph Oldam
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Irina Tchernyshyov
- Advanced Clinical Biosystems Institute, Cedars-Sinai Hospital, Beverly Hills, CA, United States
| | - Jennifer Van Eyk
- Advanced Clinical Biosystems Institute, Cedars-Sinai Hospital, Beverly Hills, CA, United States
| | - Juan Troncoso
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Charles G. Glabe
- University of California Irvine School of Biological Sciences, Irvine, CA, United States
| | - Giulio Agnetti
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| |
Collapse
|
6
|
İnci A, Dökmeci S. Extracellular chaperones in lysosomal storage diseases. Mol Genet Metab 2025; 145:109086. [PMID: 40106871 DOI: 10.1016/j.ymgme.2025.109086] [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: 11/27/2024] [Revised: 02/23/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Lysosomal storage disorders (LSDs) are a diverse group of inherited metabolic disorders characterized by the accumulation of undegraded substrates within lysosomes due to defective lysosomal function. Recent research has highlighted the pivotal role of extracellular chaperones in the pathophysiology of LSDs, revealing their crucial involvement in modulating disease progression. These chaperones aid in stabilizing and refolding misfolded lysosomal enzymes, enhancing their proper trafficking and function, which in turn reduces substrate accumulation. Furthermore, extracellular chaperones have emerged as promising biomarkers, with their levels in bodily fluids offering potential for disease diagnosis and monitoring. This review explores the current understanding of extracellular chaperones in the context of LSDs, examining their mechanisms of action, biomarker and therapeutic potential, and future directions in clinical application of LSDs.
Collapse
Affiliation(s)
- Aslı İnci
- Gazi University School of Medicine, Department of Pediatric Metabolism, Ankara, Turkey; Hacettepe University School of Medicine, Department of Medical Biology, Ankara, Turkey.
| | - Serap Dökmeci
- Hacettepe University School of Medicine, Department of Medical Biology, Ankara, Turkey
| |
Collapse
|
7
|
Luyckx T, Grootaert C, Delcour JA, Housmans JAJ, Rousseau F, Schymkowitz J, Carpentier S, Van Camp J. Impact of amyloid-like ovalbumin fibril consumption on health-related markers: An in vitro approach. Food Res Int 2025; 208:116288. [PMID: 40263866 DOI: 10.1016/j.foodres.2025.116288] [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: 11/01/2024] [Revised: 02/16/2025] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
Induction of amyloid-like morphology in food proteins offers high potential to induce new techno-functional properties in food products (e.g. use as emulsifier, thickener or gelling agent in e.g. bakery and confectionery products). However, the health impact of amyloid-like fibril (ALF) consumption remains widely understudied and merits additional research. The aim of this study was to (partially) elucidate the general health impact of food-borne ALF consumption, using egg white ovalbumin as a case study. Based on in vitro cell culture models it was demonstrated that ovalbumin ALFs (i) do not induce direct cytotoxic effects on intestinal (Caco-2, IPEC-J2) and neuronal (SH-SY5Y) cell lines, but (ii) are able to induce a Toll-like-receptor-mediated innate immune response, similar to endogenous amyloids, in activated THP-1 cells. Furthermore, the consecutive in vitro digestion and absorption (enterocyte and M-cell) experiments demonstrated that ovalbumin ALFs (i) do not completely lose their ALF morphology upon in vitro gastrointestinal digestion, and that (ii) the ALF core sequences, located at the center of the ALF structure, are transported across Caco-2 based cell models, suggesting aggregate transport. In vivo, intestinal translocation of ingested ALFs would imply potential cross-seeding of endogenous, disease-related precursor proteins. The ability of ovalbumin ALFs to induce aggregation of a disease-related precursor protein, αSyn, was evaluated in a precursor overexpressing cell model. Here, it was illustrated that only homologous (αSyn) - but not heterologous (ovalbumin) - seeding resulted in intracellular aggregation bodies of (phosphorylated) αSyn. The lack of cross-seeding supports the assumption that ovalbumin ALF consumption is not a risk factor for the development of α-synucleinopathies like Parkinson's disease.
Collapse
Affiliation(s)
- Trui Luyckx
- Laboratory of Food Chemistry and Human Nutrition, Ghent University, Ghent, Belgium
| | - Charlotte Grootaert
- Laboratory of Food Chemistry and Human Nutrition, Ghent University, Ghent, Belgium
| | - Jan A Delcour
- Laboratory of Food Chemistry and Biochemistry, KU Leuven, Leuven, Belgium
| | - Joëlle A J Housmans
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium; Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Sebastien Carpentier
- Facility for Systems Biology Based Mass Spectrometry, KU Leuven, Leuven, Belgium
| | - John Van Camp
- Laboratory of Food Chemistry and Human Nutrition, Ghent University, Ghent, Belgium.
| |
Collapse
|
8
|
Vanherle S, Janssen A, Gutiérrez de Ravé M, Janssen B, Lodder C, Botella Lucena P, Kessels S, Hardy J, Vandeput E, Wang Y, Stancu IC, Segal A, Kleinewietfeld M, Voets T, Brône B, Poovathingal S, Alpizar YA, Dewachter I. APOE deficiency inhibits amyloid-facilitated (A) tau pathology (T) and neurodegeneration (N), halting progressive ATN pathology in a preclinical model. Mol Psychiatry 2025:10.1038/s41380-025-03036-7. [PMID: 40307424 DOI: 10.1038/s41380-025-03036-7] [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: 07/03/2024] [Revised: 03/12/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025]
Abstract
In AD, amyloid pathology (A) precedes progressive development of tau pathology (T) and neurodegeneration (N), with the latter (T/N) processes associated with symptom progression. Recent anti-amyloid beta (Aβ) clinical trials raise hope but indicate the need for multi-targeted therapies, to effectively halt clinical AD and ATN pathology progression. APOE-related putative protective mutations (including APOE3Christchurch, RELN-COLBOS) were recently identified in case reports with exceptionally high resilience to autosomal dominant AD. In these cases, Nature provided proof of concept for halting autosomal dominant AD and ATN progression in humans, despite a high amyloid load, and pointing to the APOE pathway as a potential target. This is further supported by the recent identification of APOE4 homozygosity as genetic AD. Here we studied the role of APOE in a preclinical model that robustly mimics amyloid-facilitated (A) tau pathology (T) and subsequent neurodegeneration (N), denoted as ATN model, generated by crossing 5xFAD (F +) and TauP301S (T +) mice. We show that APOE deficiency, markedly inhibited progression to tau pathology and tau-induced neurodegeneration in this ATN model, despite a high Aβ load, reminiscent of the high resilience ADAD case reports. Further study identified, despite increased Aβ load (W02 stained), a significant decrease in compacted, dense core plaques stained by ThioS in APOE deficient ATN mice. Furthermore, single-cell RNA sequencing (scRNA-seq) showed a crucial role of APOE in microglial conversion beyond homeostatic microglia to reactive and disease associated microglia (DAM) in this ATN preclinical model. Microglial elimination significantly decreased amyloid-driven tau pathology, in the presence of APOE, but not in APOE deficient mice. Together the data demonstrate that APOE deficiency inhibits amyloid-driven tau pathology and subsequent neurodegeneration, by pleiotropic effects including prevention of dense core plaque formation and halting conversion of homeostatic microglia. We here present a model recapitulating inhibition of amyloid-facilitated tau pathology by APOE deficiency despite high Aβ load, important for understanding the role of APOE, and APOE-dependent processes in ATN progression and its therapeutic exploitation in AD.
Collapse
Affiliation(s)
- Sarah Vanherle
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Art Janssen
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Manuel Gutiérrez de Ravé
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Bieke Janssen
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Chritica Lodder
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Pablo Botella Lucena
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Sofie Kessels
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Jana Hardy
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Eline Vandeput
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Yanyan Wang
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Ilie-Cosmin Stancu
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | - Andrei Segal
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Bert Brône
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
| | | | - Yeranddy A Alpizar
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Brain & Disease Research, Leuven, Belgium
| | - Ilse Dewachter
- Department of Neurosciences, Biomedical Research Institute BIOMED, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
9
|
Desai AA, Zupancic JM, Trzeciakiewicz H, Gerson JE, DuBois KN, Skinner ME, Sharkey LM, McArthur N, Ferris SP, Bhatt NN, Makowski EK, Smith MD, Chen H, Huang J, Jerez C, Kuo YH, Kane RS, Kanaan NM, Paulson HL, Tessier PM. Facile generation of drug-like conformational antibodies specific for amyloid fibrils. Nat Chem Biol 2025:10.1038/s41589-025-01881-9. [PMID: 40301692 DOI: 10.1038/s41589-025-01881-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 03/11/2025] [Indexed: 05/01/2025]
Abstract
Antibodies that recognize insoluble antigens, such as amyloid fibrils associated with neurodegenerative disorders, are important for research, diagnostic and therapeutic applications. However, these types of antibodies are difficult to generate, typically require animal immunization and also commonly require humanization in the case of therapeutic applications. Here we report a methodology for generating high-quality, fully human, conformation-specific antibodies against amyloid fibrils using a published human nonimmune library, yeast-surface display and quantitative fluorescence-activated cell sorting. Notably, this approach enables the isolation of conformation-specific antibodies against tau fibrils (Alzheimer's disease) and α-synuclein fibrils (Parkinson's disease) with combinations of high affinity, high conformational specificity and, in some cases, low off-target binding that rival or exceed those of clinical-stage antibodies specific for tau (zagotenemab) and α-synuclein (cinpanemab). This approach is expected to simplify the generation of conformation-specific antibodies against diverse protein aggregates and other insoluble antigens.
Collapse
Affiliation(s)
- Alec A Desai
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jennifer M Zupancic
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Hanna Trzeciakiewicz
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Julia E Gerson
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Kelly N DuBois
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
| | - Mary E Skinner
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Lisa M Sharkey
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Nikki McArthur
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sean P Ferris
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Nemil N Bhatt
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Emily K Makowski
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Matthew D Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Hongwei Chen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Jie Huang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, TX, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yun-Huai Kuo
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ravi S Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, USA
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, MI, USA
- Michigan Alzheimer's Disease Center, University of Michigan, Ann Arbor, MI, USA
| | - Peter M Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, MI, USA.
- Michigan Alzheimer's Disease Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Randhawa S, Saini TC, Bathla M, Bhardwaj R, Dhiman R, Acharya A. Nanomaterials in targeting amyloid-β oligomers: current advances and future directions for Alzheimer's disease diagnosis and therapy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2025; 16:561-580. [PMID: 40297247 PMCID: PMC12035877 DOI: 10.3762/bjnano.16.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
The amyloid cascade hypothesis posits that amyloid-β oligomers (AβOs) are the most neurotoxic species in Alzheimer's disease (AD). These oligomers, characterized by their high β-sheet content, have been shown to significantly disrupt cell membranes, induce local inflammation, and impair autophagy processes, which collectively contribute to neuronal loss. As such, targeting AβOs specifically, rather than solely focusing on amyloid-β fibrils (AβFs), may offer a more effective therapeutic approach for AD. Recent advances in detection and diagnosis have emphasized the importance of accurately identifying AβOs in patient samples, enhancing the potential for timely intervention. In recent years, nanomaterials (NMs) have emerged as promising agents for addressing AβOs regarding their multivalent interactions, which can more effectively detect and inhibit AβO formation. This review provides an in-depth analysis of various nanochaperones developed to target AβOs, detailing their mechanisms of action and therapeutic potential via focusing on two main strategies, namely, disruption of AβOs through direct interaction and the inhibition of AβO nucleation by binding to intermediates of the oligomerization process. Evidence from in vivo studies indicate that NMs hold promise for ameliorating AD symptoms. Additionally, the review explores the different interaction mechanisms through which nanoparticles exhibit their inhibitory effects on AβOs, providing insights into their potential for clinical application. This comprehensive overview highlights the current advancements in NM-based therapies for AD and outlines future research directions aimed at optimizing these innovative treatments.
Collapse
Affiliation(s)
- Shiwani Randhawa
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Trilok Chand Saini
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Manik Bathla
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rahul Bhardwaj
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Rubina Dhiman
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
| | - Amitabha Acharya
- Biotechnology Division, C.S.I.R – Institute of Himalayan Bioresource Technology, Palampur, Himachal Prasesh, 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
11
|
Chen Y, Yin J, Liu Y, Huang Y, Zong W, Tan R. Molecular mechanism of the effect of ZnCl 2 and MgCl 2 solution on the conformation of the tau 267-312 monomer. SOFT MATTER 2025; 21:3092-3100. [PMID: 40165595 DOI: 10.1039/d4sm01546k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Alzheimer's disease is generally believed to be caused by abnormal aggregation of tau protein; however, there remains a lack of understanding about the aggregation process of tau protein in a solution environment. To explore the conformational properties of the tau protein monomer (tau267-312) in the presence of zinc and magnesium ions, we performed all-atom molecular dynamics simulations of tau267-312 in solutions of zinc chloride and magnesium chloride at different concentrations and compared these results with those obtained in pure water. The calculation results show that the β-sheet content increases significantly in the presence of zinc and magnesium ions, which causes a more compact structure for the tau protein monomers. Furthermore, it was found that stronger interactions between residues, as well as alterations in hydrophilic and hydrophobic interactions, are molecular mechanisms driving structural changes within the tau protein monomers. These findings suggest that zinc and magnesium ions facilitate a more stable conformation and promote the aggregation of tau protein monomers, which is important for understanding the aggregation and folding process of tau protein in the environment of saline solution.
Collapse
Affiliation(s)
- Yipeng Chen
- Department of Physics, Jiangxi Science and Technology Normal University, Nanchang, 330038, China.
| | - Jiantao Yin
- Department of Physics, Jiangxi Science and Technology Normal University, Nanchang, 330038, China.
| | - Yanhui Liu
- College of Physics, Guizhou University, Guiyang, 550025, China
| | - Yue Huang
- Department of Physics, Jiangxi Science and Technology Normal University, Nanchang, 330038, China.
| | - Wenjun Zong
- Department of Physics, Jiangxi Science and Technology Normal University, Nanchang, 330038, China.
| | - Rongri Tan
- Department of Physics, Jiangxi Science and Technology Normal University, Nanchang, 330038, China.
| |
Collapse
|
12
|
Roterman I, Słupina M, Dułak D, Konieczny L. Three scenarios for amyloid transformation in the context of the funnel model. Comput Struct Biotechnol J 2025; 27:1648-1659. [PMID: 40330620 PMCID: PMC12053986 DOI: 10.1016/j.csbj.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Analysis of the structure of proteins based on the evaluation of their geometric structure (secondary and supersecondary structure) can be extended to assess the structuring of the hydrophobic region of a protein. Such analysis of amyloid protein structures leads to the identification of three scenarios for amyloid formation. One is the loss of the micelle-like ordering present in the native form (a centric hydrophobic nucleus with a polar surface) in favour of a disordered distribution of hydrophobicity in the amyloid form. The term "micelle-like" is to be understood as specific hydrophobic burial. The second scenario is the reverse process, when the highly disordered distribution of hydrophobicity in the native form is replaced by a hydrophobic burial after amyloid transformation. These two scenarios have been identified for pathological (neurodegenerative) amyloids. The third scenario is the presence of hydrophobic burial ordering in a functional amyloid fibril. In this case, this ordering is present both in the fibril and in the single chain that is the building block of the fibril. This hydrophobic burial ordering provides a means of self-control of fibril size. It prevents unrestricted fibril propagation, which in the case of pathological amyloids is the main factor that disrupts the normal functioning of organelles in the amyloid surroundings. Population analysis (including numerous polymorphic forms) was performed using a collection of structures deposited in the Amyloid Atlas database. These observations allow the construction of a kind of amyloid funnel model, in which the energy minimum depends on external, environmental conditions that may be evaluated using the fuzzy oil drop model in its modified version (FOD-M).
Collapse
Affiliation(s)
- I. Roterman
- Department of Bioinformatics and Telemedicine, Jagiellonian University Medical College, ul. Medyczna 7, Kraków 30-688, Poland
| | - M. Słupina
- ALSTOM ZWUS Sp. z o.o., Modelarska 12, Katowice 40-142, Poland
| | - D. Dułak
- ASSA ABLOY Opening Solutions, Magazynowa 4, Leszno, Poland
| | - L. Konieczny
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul. Kopernika 7, Kraków 31-034, Poland
| |
Collapse
|
13
|
Sahoo S, Bandyopadhyay S. Exploring the structure and stability of pentameric amyloid β peptide aggregates in aqueous ammonium-based ionic liquid solutions. Phys Chem Chem Phys 2025; 27:7650-7664. [PMID: 40145365 DOI: 10.1039/d4cp04284k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
The self-assembly of amyloid beta (Aβ) proteins into fibrils is linked to Alzheimer's disease (AD). Soluble pentamers, particularly those formed in the early stages of Aβ aggregation, are considered highly neurotoxic. This study uses molecular dynamics simulations to explore how trimethylammonium chloride (TMAC), cholinium chloride (ChoC), and tetrabutylammonium chloride (TBAC) ionic liquids (ILs) affect the conformational stability and the association mechanism of Aβ pentameters. These ILs, characterized by varying hydrophilicity/hydrophobicity, exert differential effects on the conformatioanl flexibility of Aβ pentameters. Computational analyses reveal that TBAC induces greater conformational flexibility and multiple energetically favorable states for the Aβ pentamer, potentially driving the pentamerization process along various pathways to form different polymorphic Aβ fibrillar structures. Moreover, analysis of solvent distributions demonstrates that exchange of water by IL ion pairs at the pentamer's exterior surface primarily occurs beyond the first layer of surface-bound water molecules. Particularly, hydrophobic TBA cations show an enhanced propensity to replace weakly interacting water molecules on the surface. Mechanistic insights derived from umbrella sampling simulations further elucidate how ILs modulate the association/dissociation of Aβ monomers within pentameric aggregates. Our findings indicate that the binding of the Aβ peptide becomes less favorable and the binding free energy decreases when transitioning from TMAC to TBAC solutions, as compared to a pure aqueous solution. Finally, energy landscape analysis of Aβ peptide docking to Aβ pentameters reveals multiple low-energy conformations, which are more dispersed in the presence of ChoC and TBAC solutions, potentially hindering Aβ prefibril growth.
Collapse
Affiliation(s)
- Subhadip Sahoo
- Centre for Computational and Data Sciences, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
| | - Sanjoy Bandyopadhyay
- Molecular Modeling Laboratory, Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur-721302, India.
| |
Collapse
|
14
|
Hornung S, Vogl DP, Naltsas D, Volta BD, Ballmann M, Marcon B, Syed MMK, Wu Y, Spanopoulou A, Feederle R, Heidrich L, Bernhagen J, Koeglsperger T, Höglinger GU, Rammes G, Lashuel HA, Kapurniotu A. Multi-Targeting Macrocyclic Peptides as Nanomolar Inhibitors of Self- and Cross-Seeded Amyloid Self-Assembly of α-Synuclein. Angew Chem Int Ed Engl 2025; 64:e202422834. [PMID: 39822034 DOI: 10.1002/anie.202422834] [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: 11/23/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Amyloid self-assembly of α-synuclein (αSyn) is linked to the pathogenesis of Parkinson's disease (PD). Type 2 diabetes (T2D) has recently emerged as a risk factor for PD. Cross-interactions between their amyloidogenic proteins may act as molecular links. In fact, fibrils of islet amyloid polypeptide (IAPP) (T2D) can cross-seed αSyn amyloidogenesis and αSyn and IAPP colocalize in PD brains. Inhibition of both self- and IAPP-cross-seeded αSyn amyloidogenesis could thus interfere with PD pathogenesis. Here we show that macrocyclic peptides, designed to mimic IAPP self-/cross-interaction sites and previously found to inhibit amyloidogenesis of IAPP and/or Alzheimer's disease (AD) amyloid-β peptide Aβ40(42), are nanomolar inhibitors of both self- and IAPP-cross-seeded amyloid self-assembly of αSyn. Anti-amyloid function is mediated by nanomolar affinity interactions with αSyn via three αSyn regions which are identified as key sites of both αSyn self-assembly and its cross-interactions with IAPP. We also show that the peptides block Aβ42-mediated cross-seeding of αSyn as well. Based on their broad spectrum anti-amyloid function and additional drug-like features, these peptides are leads for multifunctional anti-amyloid drugs in PD, T2D, AD, and their comorbidities, while the identified αSyn key segments are valuable targets for novel, multi-site targeting amyloid inhibitors in PD and related synucleinopathies.
Collapse
Affiliation(s)
- Simon Hornung
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| | - Dominik P Vogl
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
- Current address, Boehringer Ingelheim, Vienna, Austria
| | - Denise Naltsas
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| | - Beatrice Dalla Volta
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| | - Markus Ballmann
- Department of Anesthesiology and Intensive Care, Technische Universität München (TUM)/, Klinikum Rechts der Isar, Ismaningerstr. 22, D-81675, Munich, Germany
| | - Beatrice Marcon
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| | - Muhammed Muazzam Kamil Syed
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, École Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Lausanne, Switzerland
| | - Yiyang Wu
- Department of Neurology, LMU University Hospital, Ludwig-Maximilian-University (LMU), Marchioninistr. 15, D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Anna Spanopoulou
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
- Current address, ITM Isotope Technologies Munich SE, Garching/Munich, Germany
| | - Regina Feederle
- Core Facility Monoclonal Antibodies, Helmholtz Center Munich German Research Center for Environmental Health, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Luzia Heidrich
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
- Current address, Life & Brain GmbH, Bonn, Germany
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilian-University (LMU), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Thomas Koeglsperger
- Department of Neurology, LMU University Hospital, Ludwig-Maximilian-University (LMU), Marchioninistr. 15, D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Günter U Höglinger
- Department of Neurology, LMU University Hospital, Ludwig-Maximilian-University (LMU), Marchioninistr. 15, D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, D-81377, Munich, Germany
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care, Technische Universität München (TUM)/, Klinikum Rechts der Isar, Ismaningerstr. 22, D-81675, Munich, Germany
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, École Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Lausanne, Switzerland
| | - Aphrodite Kapurniotu
- Division of Peptide Biochemistry, TUM School of Life Sciences, Technische Universität München (TUM), Emil-Erlenmeyer-Forum 5, D-85354, Freising, Germany
| |
Collapse
|
15
|
Moustouka C, Makhatadze GI. Quantitative detection of amyloid fibrils using fluorescence resonance energy transfer between engineered yellow and cyan proteins. Protein Sci 2025; 34:e70094. [PMID: 40099825 PMCID: PMC11915345 DOI: 10.1002/pro.70094] [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: 12/14/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
Over 20 human diseases are caused by or associated with amyloid formation. Developing diagnostic tools to understand the process of amyloid fibril formation is essential for creating therapeutic agents to combat these widespread and growing health problems. Here, we capitalize on our recent striking discovery that green fluorescent protein (GFP), one of the most-used proteins in molecular and cell biology, has a high intrinsic binding affinity to various structural intermediates along the fibrillation pathway, independent of amyloid sequence. Using engineered GFP with the fluorescence properties of Aquamarine and mCitrine, we developed a fluorescence resonance energy transfer (FRET)-based sensor to quantitatively monitor amyloid fibrils. The proof-of-principle characterization was performed on a test system consisting of PAPf39 fibrils.
Collapse
Affiliation(s)
- Caitlyn Moustouka
- Department of Biological SciencesRensselaer Polytechnic InstituteTroyNew YorkUSA
- Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroyNew YorkUSA
| | - George I. Makhatadze
- Department of Biological SciencesRensselaer Polytechnic InstituteTroyNew YorkUSA
- Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroyNew YorkUSA
- Department on Chemistry and Chemical BiologyRensselaer Polytechnic InstituteTroyNew YorkUSA
| |
Collapse
|
16
|
Scelsi HF, Close EGS, Huard DJE, Dunn E, Bogdanović N, Mudiyanselage SHW, Grant A, Stagg SM, Schmidt‐Krey I, Van Horn WD, Lieberman RL. Detection of non-native species formed during fibrillization of the myocilin olfactomedin domain. Protein Sci 2025; 34:e70063. [PMID: 40095382 PMCID: PMC11912428 DOI: 10.1002/pro.70063] [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: 11/15/2024] [Revised: 01/10/2025] [Accepted: 01/31/2025] [Indexed: 03/19/2025]
Abstract
Glaucoma is a group of neurodegenerative diseases that together are the leading cause of irreversible blindness worldwide. Myocilin-associated glaucoma is an inherited form of this disease, caused by intracellular aggregation of misfolded mutant myocilin. In vitro, the myocilin C-terminal olfactomedin domain (OLF), the relevant domain for glaucoma pathogenesis, can be driven to form amyloid-like fibrils under mild conditions. Here we characterize a species present during in vitro fibrillization. Purified OLF was subjected to fibrillization at concentrations required for downstream electron microscopy imaging and NMR spectroscopy. Additional biophysical techniques, including analytical ultracentrifugation and X-ray crystallography, were employed to further characterize the multicomponent mixture. Negative stain transmission electron microscopy (TEM) shows a non-native species reminiscent of known prefibrillar oligomers from other amyloid systems, NMR indicates a minor population of partially misfolded species is present in solution, and cryo-EM imaging shows two-dimensional protein arrays. The predominant soluble species remaining in solution after the fibril reaction is natively folded, as evidenced by X-ray crystallography. In summary, after incubating OLF under fibrillization-promoting conditions, there is a heterogeneous mixture consisting of soluble folded protein, mature amyloid-like fibrils, and partially misfolded intermediate species that at present belie additional molecular detail. The characterization of OLF fibrillar species illustrates the challenges associated with developing a comprehensive understanding of the fibrillization process for large, non-model amyloidogenic proteins.
Collapse
Affiliation(s)
- Hailee F. Scelsi
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Emily G. S. Close
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGeorgiaUSA
- Pacific Northwest National LaboratoryRichlandWashingtonUSA
| | - Dustin J. E. Huard
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Elijah Dunn
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Nebojša Bogdanović
- Institute of Molecular BiophysicsFlorida State UniversityTallahasseeFloridaUSA
| | - Sonali H. W. Mudiyanselage
- Biodesign Center for Personalized DiagnosticsArizona State UniversityTempeArizonaUSA
- School of Molecular SciencesArizona State UniversityTempeArizonaUSA
| | - Arshay Grant
- School of Biological SciencesGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Scott M. Stagg
- Institute of Molecular BiophysicsFlorida State UniversityTallahasseeFloridaUSA
- Department of Biological SciencesFlorida State UniversityTallahasseeFloridaUSA
| | | | - Wade D. Van Horn
- Biodesign Center for Personalized DiagnosticsArizona State UniversityTempeArizonaUSA
- School of Molecular SciencesArizona State UniversityTempeArizonaUSA
| | - Raquel L. Lieberman
- School of Chemistry and BiochemistryGeorgia Institute of TechnologyAtlantaGeorgiaUSA
| |
Collapse
|
17
|
López-García P, Tejero-Ojeda MM, Vaquero ME, Carrión-Vázquez M. Current amyloid inhibitors: Therapeutic applications and nanomaterial-based innovations. Prog Neurobiol 2025; 247:102734. [PMID: 40024279 DOI: 10.1016/j.pneurobio.2025.102734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Amyloid proteins have long been in the spotlight for being involved in many degenerative diseases including Alzheimer´s, Parkinson´s or type 2 diabetes, which currently cannot be prevented and for which there is no effective treatment or cure. Here we provide a comprehensive review of inhibitors that act directly on the amyloidogenic pathway (at the monomer, oligomer or fibril level) of key pathological amyloids, focusing on the most representative amyloid-related diseases. We discuss the latest advances in preclinical and clinical trials, focusing on cutting-edge developments, particularly on nanomaterials-based inhibitors, which offer unprecedented opportunities to address the complexity of protein misfolding disorders and are revolutionizing the landscape of anti-amyloid therapeutics. Notably, nanomaterials are impacting critical areas such as bioavailability, penetrability and functionality of compounds currently used in biomedicine, paving the way for more specific therapeutic solutions tailored to various amyloid-related diseases. Finally, we highlight the window of opportunity opened by comparative analysis with so-called functional amyloids for the development of innovative therapeutic approaches for these devastating diseases.
Collapse
|
18
|
Liu H, Zhao X, Chen J, Win YY, Cai J. Unnatural foldamers as inhibitors of Aβ aggregation via stabilizing the Aβ helix. Chem Commun (Camb) 2025; 61:4586-4594. [PMID: 40035705 PMCID: PMC11878269 DOI: 10.1039/d4cc05280c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
Protein aggregation is a critical factor in the development and progression of several human diseases, including Alzheimer's disease (AD), Huntington's disease, Parkinson's disease, and type 2 diabetes. Among these conditions, AD is recognized as the most prevalent progressive neurodegenerative disorder, characterized by the accumulation of amyloid-beta (Aβ) peptides. Neuronal toxicity is likely driven by soluble oligomeric intermediates of the Aβ peptide, which are thought to play a central role in the cascade leading to neuronal dysfunction and cognitive decline. In response, numerous therapeutic strategies have been developed to inhibit Aβ oligomerization, as this is believed to delay the formation of Aβ protofibrils. Traditional research has focused on discovering small molecules or peptides that antagonize Aβ oligomerization. However, recent studies have explored an alternative approach-developing ligands that stabilize the Aβ peptide in its α-helical conformation. This stabilization is thought to alter the peptide's natural aggregation kinetics, shifting it away from toxic oligomer formation and toward less harmful states. Crucially, by maintaining Aβ in this α-helical form, these ligands have been shown to rescue the peptide's associated cytotoxicity, offering a promising mechanism to mitigate the detrimental effects of Aβ in AD. While challenges remain, including treatment costs and side effects like ARIA (amyloid-related imaging abnormalities), anti-Aβ drug development represents a major advancement in Alzheimer's research and therapeutic options. This brief review aims to highlight the development and potential of these α-helix-stabilizing ligands as antagonists of Aβ aggregation, focusing on their interactions with Aβ and how these compounds induce and maintain secondary structural changes in the Aβ peptide. Notably, this innovative strategy holds promise beyond Aβ-related pathology, as the fundamental principles could be applied to other amyloidogenic proteins implicated in various amyloid-related diseases, potentially broadening the scope of therapeutic intervention for multiple neurodegenerative conditions.
Collapse
Affiliation(s)
- Heng Liu
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| | - Xue Zhao
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| | - Jianyu Chen
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| | - Yu Yu Win
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| |
Collapse
|
19
|
Andersson E, Lindblom N, Janelidze S, Salvadó G, Gkanatsiou E, Söderberg L, Möller C, Lannfelt L, Ge J, Hanrieder J, Blennow K, Deierborg T, Mattsson-Carlgren N, Zetterberg H, Gouras G, Hansson O. Soluble cerebral Aβ protofibrils link Aβ plaque pathology to changes in CSF Aβ 42/Aβ 40 ratios, neurofilament light and tau in Alzheimer's disease model mice. NATURE AGING 2025; 5:366-375. [PMID: 39939821 PMCID: PMC11922755 DOI: 10.1038/s43587-025-00810-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 01/09/2025] [Indexed: 02/14/2025]
Abstract
The Aβ42/Aβ40 ratio in the cerebrospinal fluid (CSF) and the concentrations of neurofilament light (NfL) and total tau (t-tau) are changed in the early stages of Alzheimer's disease (AD)1, but their neurobiological correlates are not entirely understood. Here, we used 5xFAD transgenic mice to investigate the associations between these CSF biomarkers and measures of cerebral Aβ, including Aβ42/Aβ40 ratios in plaques, insoluble fibrillar deposits and soluble protofibrils. A high Aβ42/Aβ40 ratio in soluble protofibrils was the strongest independent predictor of low CSF Aβ42/Aβ40 ratios and high CSF NfL and t-tau concentrations when compared to Aβ42/Aβ40 ratios in plaques and insoluble fibrillar deposits. Furthermore, the Aβ42/Aβ40 ratio in soluble protofibrils fully mediated the associations between the corresponding ratio in plaques and all the investigated CSF biomarkers. In AppNL-G-F/NL-G-F knock-in mice, protofibrils fully mediated the association between plaques and the CSF Aβ42/Aβ40 ratio. Together, the results suggest that the Aβ42/Aβ40 ratio in CSF might better reflect brain levels of soluble Aβ protofibrils than insoluble Aβ fibrils in plaques in AD. Furthermore, elevated concentrations of NfL and t-tau in CSF might be triggered by increased brain levels of soluble Aβ protofibrils.
Collapse
Affiliation(s)
| | - Nils Lindblom
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | | | - Gemma Salvadó
- Clinical Memory Research Unit, Lund University, Lund, Sweden
| | | | | | | | - Lars Lannfelt
- BioArctic AB, Stockholm, Sweden
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, Sweden
| | - Junyue Ge
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Gunnar Gouras
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Lund University, Lund, Sweden.
- Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
20
|
Urbanek A, Garland EF, Prescott EE, King MC, Olerinyova A, Wareing HE, Georgieva N, Bradshaw EL, Tzokov SB, Knight A, Tartakovskii AI, Malm T, Highley JR, De S. Molecular Determinants of Protein Pathogenicity at the Single-Aggregate Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410229. [PMID: 39804980 PMCID: PMC11884545 DOI: 10.1002/advs.202410229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/06/2024] [Indexed: 01/16/2025]
Abstract
Determining the structure-function relationships of protein aggregates is a fundamental challenge in biology. These aggregates, whether formed in vitro, within cells, or in living organisms, present significant heterogeneity in their molecular features such as size, structure, and composition, making it difficult to determine how their structure influences their functions. Interpreting how these molecular features translate into functional roles is crucial for understanding cellular homeostasis and the pathogenesis of various debilitating diseases like Alzheimer's and Parkinson's. In this study, a bottom-up approach is introduced to explore how variations in protein aggregates' size, composition, post-translational modifications and point mutations profoundly influence their biological functions. Applying this method to Alzheimer's and Parkinson's associated proteins, novel disease-relevant pathways are uncovered, demonstrating how subtle alterations in composition and morphology can shift the balance between healthy and pathological states. This findings provide deeper insights into the molecular basis of protein's functions at the single-aggregate level, enhancing the knowledge of their roles in health and disease.
Collapse
Affiliation(s)
- Agnieszka Urbanek
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Emma F. Garland
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Emily E. Prescott
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Marianne C. King
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Anna Olerinyova
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Hollie E. Wareing
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Nia Georgieva
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Ellie L. Bradshaw
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Svetomir B. Tzokov
- Cryo‐Electron Microscopy Facility, School of BiosciencesUniversity of SheffieldSheffieldS10 2TNUK
| | - Alexander Knight
- Department of Physics and AstronomyUniversity of SheffieldSheffieldS3 7RHUK
| | | | - Tarja Malm
- A.I. Virtanen Institute for Molecular SciencesUniversity of Eastern FinlandKuopio70211Finland
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| | - Suman De
- Sheffield Institute for Translational Neuroscience, Division of NeuroscienceUniversity of SheffieldSheffieldS10 2HQUK
- Neuroscience InstituteUniversity of SheffieldSheffieldS10 2TNUK
| |
Collapse
|
21
|
Tang J, Wang F, Xu Z, Zou Y, Zhang Q. Mechanistic Insights into the Inhibitory and Destabilizing Effects of K353 Acetylation on Tau Peptides and Protofibrils. J Phys Chem B 2025; 129:2201-2214. [PMID: 39950843 DOI: 10.1021/acs.jpcb.4c07977] [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: 02/28/2025]
Abstract
Misfolding and aggregation of microtubule-associated tau protein is implicated in a variety of neurodegenerative disorders (named tauopathies), including Alzheimer's disease (AD) and chronic traumatic encephalopathy (CTE). AD is the most common type of dementia associated with aging, and CTE is a special tauopathy that mostly affects contact sports athletes (such as those active in American football and boxing). Experimental studies have found that tau acetylated on residue K353 exhibited a declined aggregation propensity; however, the underlying molecular mechanism remains elusive. In this study, we performed replica exchange and conventional molecular dynamics simulations of acetylated and unacetylated tau protein models in an explicit solvent. Our results revealed that the acetylated R4 (the fourth microtubule-binding repeat domain) dimer showed less β structure and more disordered conformations than the unacetylated one. K353 acetylation weakened peptide-peptide interactions and interrupted the salt-bridge network, thus inhibiting R4 dimerization. Besides, K353 acetylation reduced the β-sheet structure probability and induced loosely packed conformations of R3-R4 (the third and fourth microtubule-binding repeat regions) protofibrils. The replacement of the charged group by acyl on K353 resulted in the loss of K353-D358 salt bridges, leading to the enlargement of the β6-β7 angle and the distance between the carboxyl-terminal and β-turn region, finally eliciting an opened "H" configuration. Our work provided a clear picture of the inhibitory mechanisms of K353 acetylation on tau at the microscopic level, which may be helpful in the development of new therapeutics against tauopathies from the perspective of post-translational modification (PTMs).
Collapse
Affiliation(s)
- Jiaxing Tang
- School of Physical Education, Xiangnan University, 889 Chenzhou Avenue, Chenzhou 423000, Hunan, People's Republic of China
| | - Feng Wang
- School of Physical Education, Xiangnan University, 889 Chenzhou Avenue, Chenzhou 423000, Hunan, People's Republic of China
| | - Zhengdong Xu
- Department of Physical Education, Shanghai University of Engineering Science, 333 Long Teng Road, Shanghai 201620, People's Republic of China
| | - Yu Zou
- Department of Sport and Exercise Science, College of Education, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Qingwen Zhang
- School of Physical Education, Shanghai University of Sport, 399 Changhai Road, Shanghai 200438, People's Republic of China
| |
Collapse
|
22
|
Aubrey LD, Radford SE. How is the Amyloid Fold Built? Polymorphism and the Microscopic Mechanisms of Fibril Assembly. J Mol Biol 2025:169008. [PMID: 39954780 DOI: 10.1016/j.jmb.2025.169008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
For a given protein sequence, many, up to sometimes hundreds of different amyloid fibril folds, can be formed in vitro. Yet, fibrils extracted from, or found in, human tissue, usually at the end of a long disease process, are often structurally homogeneous. Through monitoring of amyloid assembly reactions in vitro, the scientific community has gained a detailed understanding of the kinetic mechanisms of fibril assembly and the rates at which the different processes involved occur. However, how this kinetic information relates to the structural changes as a protein transforms from its initial, native structure to the canonical cross-β structure of amyloid remain obscure. While cryoEM has yielded a plethora of high-resolution information that portray a vast variety of fibril structures, there remains little knowledge of how and why each particular structure resulted. Recent work has demonstrated that fibril structures can change over an assembly time course, despite the different fibril types having similar thermodynamic stability. This points to kinetic control of the fibrils formed, with structures that initiate or elongate faster becoming the dominant products of assembly. Annotating kinetic assembly mechanisms alongside structural analysis of the fibrils formed is required to truly understand the molecular mechanisms of amyloid formation. However, this is a complicated task. In this review, we discuss how embracing this challenge could open new frontiers in amyloid research and new opportunities for therapy.
Collapse
Affiliation(s)
- Liam D Aubrey
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Science, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Science, University of Leeds, Leeds LS2 9JT, United Kingdom.
| |
Collapse
|
23
|
Smadja DM, Abreu MM. Hyperthermia and targeting heat shock proteins: innovative approaches for neurodegenerative disorders and Long COVID. Front Neurosci 2025; 19:1475376. [PMID: 39967803 PMCID: PMC11832498 DOI: 10.3389/fnins.2025.1475376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Neurodegenerative diseases (NDs) and Long COVID represent critical and growing global health challenges, characterized by complex pathophysiological mechanisms including neuronal deterioration, protein misfolding, and persistent neuroinflammation. The emergence of innovative therapeutic approaches, such as whole-body hyperthermia (WBH), offers promising potential to modulate underlying pathophysiological mechanisms in NDs and related conditions like Long COVID. WBH, particularly in fever-range, enhances mitochondrial function, induces heat shock proteins (HSPs), and modulates neuroinflammation-benefits that pharmacological treatments often struggle to replicate. HSPs such as HSP70 and HSP90 play pivotal roles in protein folding, aggregation prevention, and cellular protection, directly targeting pathological processes seen in NDs like Alzheimer's, Parkinson's, and Huntington's disease. Preliminary findings also suggest WBH's potential to alleviate neurological symptoms in Long COVID, where persistent neuroinflammation and serotonin dysregulation are prominent. Despite the absence of robust clinical trials, the therapeutic implications of WBH extend to immune modulation and the restoration of disrupted physiological pathways. However, the dual nature of hyperthermia's effects-balancing pro-inflammatory and anti-inflammatory responses-emphasizes the need for dose-controlled applications and stringent patient monitoring to minimize risks in vulnerable populations. While WBH shows potential interest, significant challenges remain. These include individual variability in response, limited accessibility to advanced hyperthermia technologies, and the need for standardized clinical protocols. Future research must focus on targeted clinical trials, biomarker identification, and personalized treatment strategies to optimize WBH's efficacy in NDs and Long COVID. The integration of WBH into therapeutic paradigms could mark a transformative step in addressing these complex conditions.
Collapse
Affiliation(s)
- David M. Smadja
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Hematology Department, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Paris, France
| | - M. Marc Abreu
- BTT Medical Institute, Aventura, FL, United States
- BTT Engineering Department, Aventura, FL, United States
| |
Collapse
|
24
|
Nidamangala Srinivasa A, Campbell S, Venkatesan S, Nuckolls NL, Lange JJ, Halfmann R, Zanders SE. Functional constraints of wtf killer meiotic drivers. PLoS Genet 2025; 21:e1011534. [PMID: 39965018 PMCID: PMC11892871 DOI: 10.1371/journal.pgen.1011534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/10/2025] [Accepted: 12/11/2024] [Indexed: 02/20/2025] Open
Abstract
Killer meiotic drivers are selfish DNA loci that sabotage the gametes that do not inherit them from a driver+/driver- heterozygote. These drivers often employ toxic proteins that target essential cellular functions to cause the destruction of driver- gametes. Identifying the mechanisms of drivers can expand our understanding of infertility and reveal novel insights about the cellular functions targeted by drivers. In this work, we explore the molecular mechanisms underlying the wtf family of killer meiotic drivers found in fission yeasts. Each wtf killer acts using a toxic Wtfpoison protein that can be neutralized by a corresponding Wtfantidote protein. The wtf genes are rapidly evolving and extremely diverse. Here we found that self-assembly of Wtfpoison proteins is broadly conserved and associated with toxicity across the gene family, despite minimal amino acid conservation. In addition, we found the toxicity of Wtfpoison assemblies can be modulated by protein tags designed to increase or decrease the extent of the Wtfpoison assembly, implicating assembly size in toxicity. We also identified a conserved, critical role for the specific co-assembly of the Wtfpoison and Wtfantidote proteins in promoting effective neutralization of Wtfpoison toxicity. Finally, we engineered wtf alleles that encode toxic Wtfpoison proteins that are not effectively neutralized by their corresponding Wtfantidote proteins. The possibility of such self-destructive alleles reveals functional constraints on wtf evolution and suggests similar alleles could be cryptic contributors to infertility in fission yeast populations. As rapidly evolving killer meiotic drivers are widespread in eukaryotes, analogous self-killing drive alleles could contribute to sporadic infertility in many lineages.
Collapse
Affiliation(s)
- Ananya Nidamangala Srinivasa
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Samuel Campbell
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Shriram Venkatesan
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Nicole L. Nuckolls
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Jeffrey J. Lange
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Randal Halfmann
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Sarah E. Zanders
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
25
|
Sahoo S, Bandyopadhyay S. Investigating the Restricted Dynamical Environment in and Around Aβ Peptide Oligomers in Aqueous Ionic Liquid Solutions. J Phys Chem B 2025; 129:1214-1228. [PMID: 39810736 DOI: 10.1021/acs.jpcb.4c07336] [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: 01/16/2025]
Abstract
It is widely believed that the aggregation of amyloid β (Aβ) peptides into soluble oligomers is the root cause behind Alzheimer's disease. In this study, we have performed room-temperature molecular dynamics (MD) simulations of aggregated Aβ oligomers of different sizes (pentamer (O(5)), decamer (O(10)), and hexadecamer (O(16))) in binary aqueous solutions containing 1-butyl-3-methylimidazolium tetrafluoroborate ([BMIM][BF4]) ionic liquid (IL). Investigations have been carried out to obtain a microscopic understanding of the effects of the IL on the dynamic environment around the exterior surfaces and within the confined nanocores of the oligomers. The calculations revealed that in contrast to nearly uniform dynamics near the exterior surface, heterogeneous structural distortions of oligomers of varying sizes and nonuniform distributions of water and IL components within their core volumes modify the core dynamics in a differential manner. It is demonstrated that increasingly restricted mobility of water and IL components is the origin behind the longer time scale of dynamic heterogeneity in and around the oligomers. Importantly, due to the equivalent nondirectional nature of the B-F bonds, BF4- anions are found to reorient on a time scale faster than that of water molecules. Further, the structural relaxation of protein-anion (PA) hydrogen bonds around the oligomers has been found to be correlated with sluggish translational motions of the anions but anticorrelated with their reorientational time scale. In addition, it is quantified that compared to the pure aqueous medium, strengthening of protein-water (PW) hydrogen bonds in the presence of the IL leads to their longer lifetimes.
Collapse
Affiliation(s)
- Subhadip Sahoo
- Centre for Computational and Data Sciences, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Sanjoy Bandyopadhyay
- Molecular Modeling Laboratory, Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
26
|
Lo Cascio F, Park S, Sengupta U, Puangmalai N, Bhatt N, Shchankin N, Jerez C, Moreno N, Bittar A, Xavier R, Zhao Y, Wang C, Fu H, Ma Q, Montalbano M, Kayed R. Brain-derived tau oligomer polymorphs: distinct aggregations, stability profiles, and biological activities. Commun Biol 2025; 8:53. [PMID: 39809992 PMCID: PMC11733013 DOI: 10.1038/s42003-025-07499-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Aggregation of microtubule-associated tau protein is a distinct hallmark of several neurodegenerative disorders such as Alzheimer's disease (AD), dementia with Lewy bodies (DLB), and progressive supranuclear palsy (PSP). Tau oligomers are suggested to be the primary neurotoxic species that initiate aggregation and propagate prion-like structures. Furthermore, different diseases are shown to have distinct structural characteristics of aggregated tau, denoted as polymorphs. Here, we investigate the structural and functional differences of amplified brain-derived tau oligomers (aBDTOs) from AD, DLB, and PSP. Our results indicate that the aBDTOs possess different structural and morphological features that impact neuronal function, gene regulation, and ultimately disease progression. The distinct tau oligomeric polymorphs may thus contribute to the development of clinical phenotypes and shape the progression of diseases. Our results can provide insight into developing personalized therapy to target a specific neurotoxic tau polymorph.
Collapse
Affiliation(s)
- Filippa Lo Cascio
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Suhyeorn Park
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Urmi Sengupta
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Nikita Shchankin
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Naomi Moreno
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Alice Bittar
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rhea Xavier
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yingxin Zhao
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Cankun Wang
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Hongjun Fu
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA.
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA.
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
27
|
Huang L, Ma L, Zhao Q, Zhu Q, She G, Mu L, Shi W. Simultaneous Imaging of pH and Peroxynitrite in the Endoplasmic Reticulum and Mitochondria: Revealing Organelle Interactions in Alzheimer's Disease Pathogenesis. Anal Chem 2025; 97:194-202. [PMID: 39723923 DOI: 10.1021/acs.analchem.4c03646] [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: 12/28/2024]
Abstract
pH and peroxynitrite (ONOO-) are two critical biomarkers to unveil the corresponding status of endoplasmic reticulum (ER) stress and mitochondrial dysfunction, which are closely related to Alzheimer's disease (AD). Simultaneously monitoring pH and ONOO- fluctuations in the ER and mitochondria during AD progression is pivotal for clarifying the interplay between the disorders of the two organelles and revealing AD pathogenesis. Herein, we designed and synthesized a dual-channel fluorescent probe (DCFP) to visualize pH and ONOO- in the ER and mitochondria. DCFP possessed excellent sensitivity and selectivity to pH and ONOO- without spectral crosstalk and was utilized in monitoring the two analytes within AD model cells and larval zebrafish. Importantly, DCFP could preferentially target mitochondria in normal cells and be enriched in the ER after mitochondrial depolarization. With the aid of DCFP, the slower acidification rate of the ER than that of mitochondria induced by Aβ oligomers (AβOs) was first identified, which could be ascribed to the relief of the AβOs-triggered ER stress through the Ca2+ migration from the ER to mitochondria. Moreover, continuous exposure to AβOs led to mitochondrial Ca2+ overload, accelerating the acidification and ONOO- overproduction within mitochondria. As a result, intracellular oxidative stress levels were elevated, further exacerbating ER stress and aggravating ER acidification in turn. The advanced understanding of the potential interplay between the ER and mitochondria in this work may offer new insights and methodologies for studying AD pathogenesis. The DCFP developed in this work could also be employed to study other diseases related to ER stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Lushan Huang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liyi Ma
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiaowen Zhao
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qichen Zhu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guangwei She
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Lixuan Mu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Wensheng Shi
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
28
|
More SR, Jha SK. Multi-Site Red-Edge Excitation Shift Reveals the Residue-Specific Solvation Dynamics during the Native to Amyloid-like Transition of an Amyloidogenic Protein. J Phys Chem B 2025; 129:176-193. [PMID: 39682034 DOI: 10.1021/acs.jpcb.4c07067] [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: 12/18/2024]
Abstract
Changes in water-protein interactions are crucial for proteins to achieve functional and nonfunctional conformations during structural transitions by modulating local stability. Amyloid-like protein aggregates in deteriorating neurons are hallmarks of neurodegenerative disorders. These aggregates form through significant structural changes, transitioning from functional native conformations to supramolecular cross-β-sheet structures via misfolded and oligomeric intermediates in a multistep process. However, the site-specific dynamics of water molecules from the native to misfolded conformations and further to oligomeric and compact amyloid structures remain poorly understood. In this study, we used the fluorescence method known as red-edge excitation shift (REES) to investigate the solvation dynamics at specific sites in various equilibrium conformations en route to the misfolding and aggregation of the functional domain of the TDP-43 protein (TDP-43tRRM). We generated three single tryptophan-single cysteine mutants of TDP-43tRRM, with the cysteines at different positions and tryptophan at a fixed position. Each sole cysteine was fluorescently labeled and used as a site-specific fluorophore along with the single tryptophan, creating four monitorable sites for REES studies. By investigating the site-specific extent of REES, we developed a residue-specific solvation dynamics map of TDP-43tRRM during its misfolding and aggregation. Our observations revealed that solvation dynamics progressively became more rigid and heterogeneous to varying extents at different sites during the transition from native to amyloid-like conformations.
Collapse
Affiliation(s)
- Sonal R More
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh Kumar Jha
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
29
|
Srinivasa AN, Campbell S, Venkatesan S, Nuckolls NL, Lange JJ, Halfmann R, Zanders SE. Functional constraints of wtf killer meiotic drivers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.27.609905. [PMID: 39677646 PMCID: PMC11642804 DOI: 10.1101/2024.08.27.609905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Killer meiotic drivers are selfish DNA loci that sabotage the gametes that do not inherit them from a driver+/driver- heterozygote. These drivers often employ toxic proteins that target essential cellular functions to cause the destruction of driver- gametes. Identifying the mechanisms of drivers can expand our understanding of infertility and reveal novel insights about the cellular functions targeted by drivers. In this work, we explore the molecular mechanisms underlying the wtf family of killer meiotic drivers found in fission yeasts. Each wtf killer acts using a toxic Wtfpoison protein that can be neutralized by a corresponding Wtfantidote protein. The wtf genes are rapidly evolving and extremely diverse. Here we found that self-assembly of Wtfpoison proteins is broadly conserved and associated with toxicity across the gene family, despite minimal amino acid conservation. In addition, we found the toxicity of Wtfpoison assemblies can be modulated by protein tags designed to increase or decrease the extent of the Wtfpoison assembly, implicating assembly size in toxicity. We also identified a conserved, critical role for the specific co-assembly of the Wtfpoison and Wtfantidote proteins in promoting effective neutralization of Wtfpoison toxicity. Finally, we engineered wtf alleles that encode toxic Wtfpoison proteins that are not effectively neutralized by their corresponding Wtfantidote proteins. The possibility of such self-destructive alleles reveals functional constraints on wtf evolution and suggests similar alleles could be cryptic contributors to infertility in fission yeast populations. As rapidly evolving killer meiotic drivers are widespread in eukaryotes, analogous self-killing drive alleles could contribute to sporadic infertility in many lineages.
Collapse
Affiliation(s)
- Ananya Nidamangala Srinivasa
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Samuel Campbell
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Shriram Venkatesan
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Nicole L. Nuckolls
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Jeffrey J. Lange
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
| | - Randal Halfmann
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Sarah E. Zanders
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
30
|
Jhawar M, Paul S. Unveiling the Inhibitory Effect of Magnolol in the Aggregation of Human Calcitonin (hCT): A Comprehensive In-Silico Study. Chemphyschem 2025; 26:e202400679. [PMID: 39432713 DOI: 10.1002/cphc.202400679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/23/2024]
Abstract
Amyloid fibril formation by some peptides leads to several neurogenetic disorders. This limits their biological activity and increases cytotoxicity. Human calcitonin (hCT), 32 residue containing peptide, known for regulating calcium and phosphate concentration in the blood tends to form amyloids in aqueous medium. Polyphenols are very effective in inhibiting fibril formation. As part of our research, we have taken Magnolol (Mag), which is extracted from the Chinese herb Magnolia officinalis. To evaluate its effectiveness as an inhibitor in preventing hCT aggregation, we conducted an all-atom classical molecular dynamics simulation with varying concentrations of Mag. In presence of Mag, hCT maintains its helical conformation in higher order. Magnolol primarily interacts with hCT via van der Waals interaction. Asp15 residue of hCT, resides in the amyloid region (D15FNKF19) forms strong hydrogen bonding interaction with Mag. Moreover, aromatic residues of hCT interact with Mag through π-π stacking interactions. Our work gives insights into the molecular mechanism of Magnolol in the inhibition of hCT fibril formation to use it as a potential candidate for medicinal purpose.
Collapse
Affiliation(s)
- Mira Jhawar
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam, 781039, India
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam, 781039, India
| |
Collapse
|
31
|
Jana AK, Güven Ö, Yaşar F. The stability and dynamics of the Aβ40/Aβ42 interlaced mixed fibrils. J Biomol Struct Dyn 2025; 43:277-290. [PMID: 37964619 DOI: 10.1080/07391102.2023.2280765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023]
Abstract
The accumulation of fibrillar amyloid-β (Aβ) aggregates in the brain, predominantly comprising 40- and 42-residue amyloid-β (Aβ40 and Aβ42), is a major pathological hallmark of Alzheimer's disease (AD). Aβ40 and Aβ42 naturally coexist in the brain under normal physiological conditions, and their interplay is generally considered to be a critical factor in the progression of AD. In addition to forming homogeneous oligomers and fibrils, Aβ40 and Aβ42 are also reported to co-assemble into hetero-oligomers and interlaced mixed fibrils, as evidenced by solid-state nuclear magnetic resonance spectroscopy (NMR), high molecular weight mass spectrometry and cross-seeding experiments. However, the exact molecular mechanisms underlying these processes remain unclear. In this study, we have used a recently resolved structurally uniform 1:1 mixture of Aβ40/Aβ42 interlaced mixed fibril as a prototype to gain insights into the molecular-level interactions between Aβ40 and Aβ42. We employed fully atomistic molecular dynamics simulation and compared the results with a homogeneous U-shaped Aβ40 fibrillar model. Our simulations using two different force fields provide conclusive evidence that the Aβ40/Aβ42 interlaced mixed fibril is energetically more favorable than the homogeneous Aβ40 fibrillar model. Furthermore, we also show that the increase in stability observed in the mixed model stems primarily from the packing interfaces and the stacking interfaces between C-termini. Our simulation results provide valuable mechanistic insights that are not readily accessible in experiment and could have significant implications for both the pathogenesis of AD and the development of current therapeutic strategies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Asis K Jana
- Department of Microbiology and Biotechnology, Sister Nivedita University, Kolkata, West Bengal, India
| | - Özgür Güven
- Department of Physics Engineering, Hacettepe University, Ankara, Türkiye
| | - Fatih Yaşar
- Department of Physics Engineering, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
32
|
Menefee K, Larios K, Rinauro DJ, Tun A, Jauregui B, Contreras JI, Nogaj LA, Moffet DA. Identifying the Role of Individual Seal IAPP Amino Acids in Inhibiting the Aggregation of Human IAPP. Protein Pept Lett 2025; 32:44-53. [PMID: 39660516 DOI: 10.2174/0109298665340227241115110404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 12/12/2024]
Abstract
INTRODUCTION The progression of type 2 diabetes in humans appears to be linked to the loss of insulin-producing β-cells. One of the major contributors to β-cell loss is the formation of toxic human IAPP amyloid (hIAPP, Islet Amyloid Polypeptide, amylin) in the pancreas. Inhibiting the formation of toxic hIAPP amyloid could slow, if not prevent altogether, the progression of type 2 diabetes. Many non-human organisms also express amyloidogenic IAPP variants known to kill pancreatic cells and give rise to diabetes-like symptoms. Surprisingly, some of these non-human IAPP variants function as inhibitors of hIAPP aggregation, raising the possibility of developing non-human IAPP peptides into anti-diabetic therapeutic peptides. One such inhibitory IAPP variant is seal IAPP, which has been shown to inhibit hIAPP aggregation. Seal IAPP only differs from hIAPP by three amino acids. In this study, each of the six seal/human IAPP permutations was analyzed to identify the role of each of the three amino acid positions in inhibiting hIAPP aggregation. AIMS This study aimed to identify the minimal amino acid substitutions to yield a peptide inhibitor of human IAPP aggregation. OBJECTIVE The goal of the study was to determine the minimal amino acid substitutions necessary to convert human IAPP into an amyloid-inhibiting peptide. METHODS The formation of toxic hIAPP amyloid was monitored using Thioflavin T binding assays, atomic force microscopy, and MTT cell rescue studies. RESULTS One seal IAPP variant retained amyloid-inhibition activity, and two variants appeared to be more amyloidogenic and toxic than wild-type human IAPP. CONCLUSION These results suggest that inhibition of hIAPP requires both the H18R and F23L substitutions of hIAPP.
Collapse
Affiliation(s)
- Kate Menefee
- Department of Chemistry and Biochemistry, Loyola Marymount University, 1 LMU Drive, Los Angeles, CA90045, USA
| | - Kelsy Larios
- Department of Biology, Mount Saint Mary's University, 12001 Chalon Drive, Los Angeles, CA90049, USA
| | - Dillon J Rinauro
- Department of Chemistry and Biochemistry, Loyola Marymount University, 1 LMU Drive, Los Angeles, CA90045, USA
| | - Angela Tun
- Department of Chemistry and Biochemistry, Loyola Marymount University, 1 LMU Drive, Los Angeles, CA90045, USA
| | - Betssy Jauregui
- Department of Chemistry and Biochemistry, Loyola Marymount University, 1 LMU Drive, Los Angeles, CA90045, USA
| | - Jessica I Contreras
- Department of Biology, Mount Saint Mary's University, 12001 Chalon Drive, Los Angeles, CA90049, USA
| | - Luiza A Nogaj
- Department of Biology, Mount Saint Mary's University, 12001 Chalon Drive, Los Angeles, CA90049, USA
| | - David A Moffet
- Department of Chemistry and Biochemistry, Loyola Marymount University, 1 LMU Drive, Los Angeles, CA90045, USA
| |
Collapse
|
33
|
Taheri F, Hou C. Life History Differences Between Lepidoptera Larvae and Blattodea Nymphs Lead to Different Energy Allocation Strategies and Cellular Qualities. INSECTS 2024; 15:991. [PMID: 39769593 PMCID: PMC11676388 DOI: 10.3390/insects15120991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025]
Abstract
Different life histories result in different strategies to allocate energy in biosynthesis, including growth and reproduction, and somatic maintenance. One of the most notable life history differences between Lepidoptera and Blattodea species is that the former grow much faster than the latter, and during metamorphosis, a large amount of tissue in Lepidoptera species disintegrates. In this review, using Lepidoptera caterpillars and cockroach nymphs as examples, we show that, due to these differences in growth processes, cockroach nymphs spend 20 times more energy on synthesizing one unit of biomass (indirect cost of growth) than butterfly caterpillars. Because of the low indirect cost of growth in caterpillars, the fraction of metabolic energy allocated to growth is six times lower, and that for maintenance is seven times higher in caterpillars, compared to cockroach nymphs, despite caterpillar's higher growth rates. Moreover, due to the higher biosynthetic energy cost in cockroach nymphs, they have better cellular qualities, including higher proteasomal activity for protein quality control and higher resistance to oxidative stress. We also show that under food restriction conditions, the fraction of assimilated energy allocated to growth was reduced by 120% in cockroach nymphs, as they lost body weight under food restriction, while this reduction was only 14% in hornworms, and the body mass increased at a lower rate. Finaly, we discuss future research, especially the difference in adult lifespans associated with the energetic differences.
Collapse
Affiliation(s)
| | - Chen Hou
- Department of Biology, Missouri University of Science and Technology, Rolla, MO 65409, USA;
| |
Collapse
|
34
|
Datta D, Navalkar A, Sakunthala A, Paul A, Patel K, Masurkar S, Gadhe L, Manna S, Bhattacharyya A, Sengupta S, Poudyal M, Devi J, Sawner AS, Kadu P, Shaw R, Pandey S, Mukherjee S, Gahlot N, Sengupta K, Maji SK. Nucleo-cytoplasmic environment modulates spatiotemporal p53 phase separation. SCIENCE ADVANCES 2024; 10:eads0427. [PMID: 39661689 PMCID: PMC11633762 DOI: 10.1126/sciadv.ads0427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Liquid-liquid phase separation of various transcription factors into biomolecular condensates plays an essential role in gene regulation. Here, using cellular models and in vitro studies, we show the spatiotemporal formation and material properties of p53 condensates that might dictate its function. In particular, p53 forms liquid-like condensates in the nucleus of cells, which can bind to DNA and perform transcriptional activity. However, cancer-associated mutations promote misfolding and partially rigidify the p53 condensates with impaired DNA binding ability. Irrespective of wild-type and mutant forms, the partitioning of p53 into cytoplasm leads to the condensate formation, which subsequently undergoes rapid solidification. In vitro studies show that abundant nuclear components such as RNA and nonspecific DNA promote multicomponent phase separation of the p53 core domain and maintain their liquid-like property, whereas specific DNA promotes its dissolution into tetrameric functional p53. This work provides mechanistic insights into how the life cycle and DNA binding properties of p53 might be regulated by phase separation.
Collapse
Affiliation(s)
- Debalina Datta
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ambuja Navalkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arunima Sakunthala
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajoy Paul
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Komal Patel
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Shalaka Masurkar
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Laxmikant Gadhe
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| | - Shouvik Manna
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Arpita Bhattacharyya
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Shinjinee Sengupta
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Manisha Poudyal
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Jyoti Devi
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ajay Singh Sawner
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Pradeep Kadu
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Ranjit Shaw
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Satyaprakash Pandey
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Semanti Mukherjee
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Nitisha Gahlot
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
| | - Kundan Sengupta
- Chromosome Biology Lab, Indian Institute of Science Education and Research, Pune, India
| | - Samir K. Maji
- Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai 400076, India
- Sunita Sanghi Centre of Aging and Neurodegenerative Diseases, IIT Bombay, Powai, Mumbai 400076, India
| |
Collapse
|
35
|
Khan MA. Targeting Iron Responsive Elements (IREs) of APP mRNA into Novel Therapeutics to Control the Translation of Amyloid-β Precursor Protein in Alzheimer's Disease. Pharmaceuticals (Basel) 2024; 17:1669. [PMID: 39770511 PMCID: PMC11677800 DOI: 10.3390/ph17121669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
The hallmark of Alzheimer's disease (AD) is the buildup of amyloid-β (Aβ), which is produced when the amyloid precursor protein (APP) misfolds and deposits as neurotoxic plaques in the brain. A functional iron responsive element (IRE) RNA stem loop is encoded by the APP 5'-UTR and may be a target for regulating the production of Alzheimer's amyloid precursor protein. Since modifying Aβ protein expression can give anti-amyloid efficacy and protective brain iron balance, targeted regulation of amyloid protein synthesis through modulation of 5'-UTR sequence function is a novel method for the prospective therapy of Alzheimer's disease. Numerous mRNA interference strategies target the 2D RNA structure, even though messenger RNAs like tRNAs and rRNAs can fold into complex, three-dimensional structures, adding even another level of complexity. The IRE family is among the few known 3D mRNA regulatory elements. This review seeks to describe the structural and functional aspects of IREs in transcripts, including that of the amyloid precursor protein, that are relevant to neurodegenerative diseases, including AD. The mRNAs encoding the proteins involved in iron metabolism are controlled by this family of similar base sequences. Like ferritin IRE RNA in their 5'-UTR, iron controls the production of APP in their 5'-UTR. Iron misregulation by iron regulatory proteins (IRPs) can also be investigated and contrasted using measurements of the expression levels of tau production, Aβ, and APP. The development of AD is aided by iron binding to Aβ, which promotes Aβ aggregation. The development of small chemical therapeutics to control IRE-modulated expression of APP is increasingly thought to target messenger RNAs. Thus, IRE-modulated APP expression in AD has important therapeutic implications by targeting mRNA structures.
Collapse
Affiliation(s)
- Mateen A Khan
- Department of Life Science, College of Science and General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
36
|
Wani NA, Gazit E, Ramamoorthy A. Interplay between Antimicrobial Peptides and Amyloid Proteins in Host Defense and Disease Modulation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:25355-25366. [PMID: 39564995 DOI: 10.1021/acs.langmuir.4c03123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
The biological properties of antimicrobial peptides (AMPs) and amyloid proteins and their cross-talks have gained increasing attention due to their potential implications in both host defense mechanisms and amyloid-related diseases. However, complex interactions, molecular mechanisms, and physiological applications are not fully understood. The interplay between antimicrobial peptides and amyloid proteins is crucial for uncovering new insights into immune defense and disease mechanisms, bridging critical gaps in understanding infectious and neurodegenerative diseases. This review provides an overview of the cross-talk between AMPs and amyloids, highlighting their intricate interplay, mechanisms of action, and potential therapeutic implications. The dual roles of AMPs, which not only serve as key components of the innate immune system, combating microbial infections, but also exhibit modulatory effects on amyloid formation and toxicity, are discussed. The diverse mechanisms employed by AMPs to modulate amyloid aggregation, fibril formation, and toxicity are also discussed. Additionally, we explore emerging evidence suggesting that amyloid proteins may possess antimicrobial properties, adding a new dimension to the intricate relationship between AMPs and amyloids. This review underscores the importance of understanding the cross-talk between AMPs and amyloids to better understand the molecular processes underlying infectious diseases and amyloid-related disorders and to aid in the development of therapeutic avenues to treat them.
Collapse
Affiliation(s)
- Naiem Ahmad Wani
- Department Chemical and Biomedical Engineering, Florida State University, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida 32310, United States
| | - Ehud Gazit
- Department of Materials Science and Engineering, Tel Aviv University, 6997801 Tel Aviv, Israel
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Ayyalusamy Ramamoorthy
- Department Chemical and Biomedical Engineering, Florida State University, Tallahassee, Florida 32310, United States
- National High Magnetic Field Laboratory, Florida State University, Tallahassee, Florida 32310, United States
- Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida 32304, United States
| |
Collapse
|
37
|
Fayoud H, Belousov MV, Antonets KS, Nizhnikov AA. Pathogenesis-Associated Bacterial Amyloids: The Network of Interactions. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2107-2132. [PMID: 39865026 DOI: 10.1134/s0006297924120022] [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: 09/13/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 01/28/2025]
Abstract
Amyloids are protein fibrils with a characteristic cross-β structure that is responsible for the unusual resistance of amyloids to various physical and chemical factors, as well as numerous pathogenic and functional consequences of amyloidogenesis. The greatest diversity of functional amyloids was identified in bacteria. The majority of bacterial amyloids are involved in virulence and pathogenesis either via facilitating formation of biofilms and adaptation of bacteria to colonization of a host organism or through direct regulation of toxicity. Recent studies have shown that, beside their commonly known activity, amyloids may be involved in the spatial regulation of proteome by modulating aggregation of other amyloidogenic proteins with multiple functional or pathological effects. Although the studies on the role of microbiome-produced amyloids in the development of amyloidoses in humans and animals have only been started, it is clear that humans as holobionts contain amyloids encoded not only by the host genome, but also by microorganisms that constitute the microbiome. Amyloids acquired from external sources (e.g., food) can interact with holobiont amyloids and modulate the effects of bacterial and host amyloids, thus adding another level of complexity to the holobiont-associated amyloid network. In this review, we described bacterial amyloids directly or indirectly involved in disease pathogenesis in humans and discussed the significance of bacterial amyloids in the three-component network of holobiont-associated amyloids.
Collapse
Affiliation(s)
- Haidar Fayoud
- Faculty of Biology, St. Petersburg State University, St. Petersburg, 199034, Russia
- All-Russia Research Institute for Agricultural Microbiology, St. Petersburg, 196608, Russia
| | - Mikhail V Belousov
- Faculty of Biology, St. Petersburg State University, St. Petersburg, 199034, Russia
- All-Russia Research Institute for Agricultural Microbiology, St. Petersburg, 196608, Russia
| | - Kirill S Antonets
- Faculty of Biology, St. Petersburg State University, St. Petersburg, 199034, Russia
- All-Russia Research Institute for Agricultural Microbiology, St. Petersburg, 196608, Russia
| | - Anton A Nizhnikov
- Faculty of Biology, St. Petersburg State University, St. Petersburg, 199034, Russia. ARRAY(0x5ae2b7af6df8)
- All-Russia Research Institute for Agricultural Microbiology, St. Petersburg, 196608, Russia
| |
Collapse
|
38
|
Budamagunta MS, Mori H, Silk J, Slez RR, Bognár B, Mendiola UR, Kálai T, Maezawa I, Voss JC. Nitroxyl Hybrids with Curcumin and Stilbene Scaffolds Display Potent Antioxidant Activity, Remodel the Amyloid Beta Oligomer, and Reverse Amyloid Beta-Induced Cytotoxicity. Antioxidants (Basel) 2024; 13:1411. [PMID: 39594552 PMCID: PMC11591036 DOI: 10.3390/antiox13111411] [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: 10/11/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The disorder and heterogeneity of low-molecular-weight amyloid-beta oligomers (AβOs) underlie their participation in multiple modes of cellular dysfunction associated with the etiology of Alzheimer's disease (AD). The lack of specified conformational states in these species complicates efforts to select or design small molecules to targeting discrete pathogenic states. Furthermore, targeting AβOs alone may be therapeutically insufficient, as AD progresses as a multifactorial, self-amplifying cascade. To address these challenges, we have screened the activity of seven new candidates that serve as Paramagnetic Amyloid Ligand (PAL) candidates. PALs are bifunctional small molecules that both remodel the AβO structure and localize a potent antioxidant that mimics the activity of SOD within live cells. The candidates are built from either a stilbene or curcumin scaffold with nitroxyl moiety to serve as catalytic antioxidants. Measurements of PAL AβO binding and remolding along with assessments of bioactivity allow for the extraction of useful SAR information from screening data. One candidate (HO-4450; PMT-307), with a six-membered nitroxyl ring attached to a stilbene ring, displays the highest potency in protecting against cell-derived Aβ. A preliminary low-dose evaluation in AD model mice provides evidence of modest treatment effects by HO-4450. The results for the curcumin PALs demonstrate that the retention of the native curcumin phenolic groups is advantageous to the design of the hybrid PAL candidates. Finally, the PAL remodeling of AβO secondary structures shows a reasonable correlation between a candidate's bioactivity and its ability to reduce the fraction of antiparallel β-strand.
Collapse
Affiliation(s)
- Madhu S. Budamagunta
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
| | - Hidetoshi Mori
- Center for Genomic Pathology, University of California Davis, Sacramento, CA 95817, USA
| | - Joshua Silk
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
- Paramag Biosciences Inc., 720 Olive Drive, Davis, CA 95616, USA
| | - Ryan R. Slez
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
| | - Balázs Bognár
- Institute of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Pécs, Honvéd St. 1., H-7624 Pécs, Hungary; (B.B.); (T.K.)
- János Szentágothai Research Center, Ifjúság St. 20., H-7624 Pécs, Hungary
| | - Ulises Ruiz Mendiola
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA;
| | - Tamás Kálai
- Institute of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Pécs, Honvéd St. 1., H-7624 Pécs, Hungary; (B.B.); (T.K.)
- János Szentágothai Research Center, Ifjúság St. 20., H-7624 Pécs, Hungary
| | - Izumi Maezawa
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA;
| | - John C. Voss
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
- Paramag Biosciences Inc., 720 Olive Drive, Davis, CA 95616, USA
| |
Collapse
|
39
|
Ho HH, Wing SS. α-Synuclein ubiquitination - functions in proteostasis and development of Lewy bodies. Front Mol Neurosci 2024; 17:1498459. [PMID: 39600913 PMCID: PMC11588729 DOI: 10.3389/fnmol.2024.1498459] [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: 09/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Synucleinopathies are neurodegenerative disorders characterized by the accumulation of α-synuclein containing Lewy bodies. Ubiquitination, a key post-translational modification, has been recognized as a pivotal regulator of α-synuclein's cellular dynamics, influencing its degradation, aggregation, and associated neurotoxicity. This review examines comprehensively the current understanding of α-synuclein ubiquitination and its role in the pathogenesis of synucleinopathies, particularly in the context of Parkinson's disease. We explore the molecular mechanisms responsible for α-synuclein ubiquitination, with a focus on the roles of E3 ligases and deubiquitinases implicated in the degradation process which occurs primarily through the endosomal lysosomal pathway. The review further discusses how the dysregulation of these mechanisms contributes to α-synuclein aggregation and LB formation and offers suggestions for future investigations into the role of α-synuclein ubiquitination. Understanding these processes may shed light on potential therapeutic avenues that can modulate α-synuclein ubiquitination to alleviate its pathological impact in synucleinopathies.
Collapse
Affiliation(s)
- Hung-Hsiang Ho
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Simon S. Wing
- Department of Medicine, McGill University and Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| |
Collapse
|
40
|
Jain M, Matysiak S. Dual Role of Anionic Lipids in Amyloid Aggregation. J Phys Chem B 2024; 128:10831-10840. [PMID: 39450869 DOI: 10.1021/acs.jpcb.4c05636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's, affect millions worldwide and share a common feature: the aggregation of intrinsically disordered proteins into toxic oligomers that interact with cell membranes. In Alzheimer's disease (AD), amyloid-beta (Aβ) peptides accumulate and bind to plasma membranes, potentially disrupting cellular function. The complex interplay between amyloidogenic peptides and lipid membranes, particularly the role of anionic lipids, is crucial in disease pathogenesis but challenging to characterize experimentally. The literature presents conflicting results on the influence of anionic lipids on peptide aggregation kinetics, highlighting a knowledge gap. To address this, we used coarse-grained molecular dynamics (CG-MD) simulations to study interactions between a model amyloidogenic peptide, amyloid-β's K16LVFFAE22 fragment (Aβ16-22), and mixed lipid bilayers. We used phosphatidylserine (PS) and phosphatidylcholine (PC) as representative anionic and zwitterionic lipids, respectively, examining the mixed bilayer compositions of 0% PS-100% PC, 10% PS-90% PC, and 30% PS-70% PC. Our simulations revealed that membranes enriched in anionic lipids enhance peptide adsorption and interaction kinetics. The aggregation dynamics was modulated by two competing factors: increased local peptide concentration near negatively charged membranes, which promoted aggregation, and peptide-lipid interactions, which slowed it down. Higher percentages of anionic lipids led to smaller and more ordered aggregates and enhanced lipid demixing, leading to the formation of PS clusters. These findings contribute to understanding membrane-mediated peptide aggregation in neurodegenerative disorders, potentially guiding new therapeutic strategies targeting the early stages of protein aggregation in various neurodegenerative diseases.
Collapse
Affiliation(s)
- Meenal Jain
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Silvina Matysiak
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland 20742, United States
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
41
|
Zarrilli B, Bonanni R, Belfiore M, Severino M, Cariati I, Fioravanti R, Cappella G, Sennato S, Frank C, Giordani C, Tancredi V, Bombelli C, Diociaiuti M, D'Arcangelo G. Molecular mechanisms at the basis of the protective effect exerted by EPPS on neurodegeneration induced by prefibrillar amyloid oligomers. Sci Rep 2024; 14:26533. [PMID: 39489758 PMCID: PMC11532462 DOI: 10.1038/s41598-024-77859-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024] Open
Abstract
It has been shown recently, without an explanation of the possible molecular mechanisms involved, that 4-(2-hydroxyethyl)-1-piperazinepropanesulphonic (EPPS) acid effectively protects from the neurotoxicity induced by oligomers and plaques formed by the protein amyloid-β protein. Here we report the same protective effect, obtained in vitro (HT22-diff cell line) and ex vivo (hippocampal slices) models, against amyloid neurotoxicity induced by oligomers of salmon Calcitonin (sCT), which has been shown to be a good model for the study of neurodegenerative diseases. Based on biophysical studies focusing on the protein aggregation kinetic and the interaction of the aggregates with model membranes, we propose a possible molecular mechanism underlying the protective effects. Taken together, our results indicate that EPPS is able to counteract the direct association (primary aggregation) of harmless low-molecular weight aggregates (dimers and trimers) or their aggregation catalysed by surfaces present in the solution (secondary aggregation). Thus, EPPS stabilizes harmless aggregates and hinders the formation of toxic and metastable prefibrillar oligomers. Overall, our data demonstrate that EPPS is an excellent drug candidate for the treatment of neurodegeneration due to misfolded proteins, such as Alzheimer's or Parkinson's disease.
Collapse
Affiliation(s)
- Beatrice Zarrilli
- Department of Systems Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
- Laboratory of Experimental Neurology, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
| | - Marcello Belfiore
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Mariagrazia Severino
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
| | - Raoul Fioravanti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Giacomo Cappella
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Sennato
- CNR - Institute of Complex Systems (ISC) - Sede "Sapienza", c/o Physics Department, "Sapienza" University of Rome, 00185, Rome, Italy
| | - Claudio Frank
- UniCamillus - Saint Camillus International University of Health Sciences, Via di Sant'Alessandro 8, Rome, 00131, Italy
| | - Cristiano Giordani
- Instituto de Física, Universidad de Antioquia, Calle 70 No. 52-21, 050010, Medellín, Colombia
- Grupo Productos Naturales Marinos, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Calle 70 No. 52-21, 050010, Medellín, Colombia
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
- Centre of Space Bio-Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
| | - Cecilia Bombelli
- CNR - Institute for Biological Systems, Secondary Office of Rome - Reaction Mechanisms c/o Chemistry Department, "Sapienza" University of Rome, Rome, Italy
| | - Marco Diociaiuti
- CNR - Institute for Biological Systems, Secondary Office of Rome - Reaction Mechanisms c/o Chemistry Department, "Sapienza" University of Rome, Rome, Italy.
| | - Giovanna D'Arcangelo
- Department of Systems Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
- Centre of Space Bio-Medicine, "Tor vergata" University of Rome, Via Montpellier 1, 00133, Rome, Italy
| |
Collapse
|
42
|
Middleton DA. NMR studies of amyloid interactions. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2024; 144-145:63-96. [PMID: 39645351 DOI: 10.1016/j.pnmrs.2024.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 12/09/2024]
Abstract
Amyloid fibrils are insoluble, fibrous nanostructures that accumulate extracellularly in biological tissue during the progression of several human disorders, including Alzheimer's disease (AD) and type 2 diabetes. Fibrils are assembled from protein monomers via the transient formation of soluble, cytotoxic oligomers, and have a common molecular architecture consisting of a spinal core of hydrogen-bonded protein β-strands. For the past 25 years, NMR spectroscopy has been at the forefront of research into the structure and assembly mechanisms of amyloid aggregates. Until the recent boom in fibril structure analysis by cryo-electron microscopy, solid-state NMR was unrivalled in its ability to provide atomic-level models of amyloid fibril architecture. Solution-state NMR has also provided complementary information on the early stages in the amyloid assembly mechanism. Now, both NMR modalities are proving to be valuable in unravelling the complex interactions between amyloid species and a diverse range of physiological metal ions, molecules and surfaces that influence the assembly pathway, kinetics, morphology and clearance in vivo. Here, an overview is presented of the main applications of solid-state and solution-state NMR for studying the interactions between amyloid proteins and biomembranes, glycosaminoglycan polysaccharides, metal ions, polyphenols, synthetic therapeutics and diagnostics. Key NMR methodology is reviewed along with examples of how to overcome the challenges of detecting interactions with aggregating proteins. The review heralds this new role for NMR in providing a comprehensive and pathologically-relevant view of the interactions between protein and non-protein components of amyloid. Coverage of both solid- and solution-state NMR methods and applications herein will be informative and valuable to the broad communities that are interested in amyloid proteins.
Collapse
Affiliation(s)
- David A Middleton
- Department of Chemistry, Lancaster University, Lancaster LA1 4YB, United Kingdom.
| |
Collapse
|
43
|
Patel SP, Nikam T, Sreepathi B, Karankar VS, Jaiswal A, Vardhan SV, Rana A, Toga V, Srivastava N, Saraf SA, Awasthi S. Unraveling the Molecular Jam: How Crowding Shapes Protein Aggregation in Neurodegenerative Disorders. ACS Chem Biol 2024; 19:2118-2130. [PMID: 39373539 DOI: 10.1021/acschembio.4c00365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Protein misfolding and aggregation are the hallmarks of neurodegenerative diseases including Huntington's disease, Parkinson's disease, Alzheimer's disease, and prion diseases. A crowded cellular environment plays a crucial role in modulating protein aggregation processes in vivo and the pathological aggregation of proteins linked to different neurodegenerative disorders. Here, we review recent studies examining the effects of various crowding agents, such as polysaccharides, polyethylene glycol, and proteins like BSA and lysozyme on the behaviors of aggregation of several amyloidogenic peptides and proteins, including amylin, huntingtin, tau, α-synuclein, prion, and amyloid-β. We also summarize how the aggregation kinetics, thermodynamic stability, and morphology of amyloid fibrils are altered significantly in the presence of crowding agents. In addition, we also discuss the molecular basis underlying the modulation of amyloidogenic aggregation, focusing on changes in the protein conformation, and the nucleation mechanism. The molecular understanding of the effects of macromolecular crowding on amyloid aggregation is essential for revealing disease pathologies and identifying possible therapeutic targets. Thus, this review offers a perspective on the complex interplay between protein aggregation and the crowded cellular environment in vivo and explains the relevance of crowding in the context of neurodegenerative disorders.
Collapse
Affiliation(s)
- Shashi Prakash Patel
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Tejas Nikam
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Bhargavi Sreepathi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Vijayshree S Karankar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Ankita Jaiswal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Salumuri Vamsi Vardhan
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Anika Rana
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Vanshu Toga
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Shubhini A Saraf
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| | - Saurabh Awasthi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Lucknow-226002, Uttar Pradesh, India
| |
Collapse
|
44
|
Sanagavarapu K, Meisl G, Lattanzi V, Bernfur K, Frohm B, Olsson U, Knowles TPJ, Malmendal A, Linse S. Serine phosphorylation mimics of Aβ form distinct, non-cross-seeding fibril morphs. Chem Sci 2024:d3sc06343g. [PMID: 39494375 PMCID: PMC11529392 DOI: 10.1039/d3sc06343g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
The self-assembly of amyloid-β peptide (Aβ) into fibrils and oligomers is linked to Alzheimer's disease (AD). Fibrillar aggregates in AD patient's brains contain several post-translational modifications, including phosphorylation at positions 8 and 26. These play a key role in modifying the aggregation propensity of Aβ, yet how they affect the mechanism of aggregation is only poorly understood. Here we elucidate the aggregation mechanism of Aβ42 peptides with phosphomimic mutations at these positions, with glutamine mimicking the size, and glutamate mimicking both the size and charge effect. We find that all variants are less aggregation-prone than wild-type Aβ42 with the glutamate mutants showing the largest reduction. Secondary nucleation is the dominant nucleation route for all variants, as confirmed using seeding experiments; however, its rate is reduced by about an order of magnitude or more for all variants relative to wild-type. S26Q and S26E fibrils fail to catalyse nucleation of wild-type monomers and vice versa, while the S8 variants co-aggregate more readily with wild-type. Ultrastructural analyses by cryo-electron microscopy and small angle X-ray scattering reveal an altered structure with longer node-to-node distance and smaller cross-section dimensions of S26Q fibrils. These results imply that structural compatibility between fibrils and monomer is a key determinant in secondary nucleation, and that small modifications can alter the preferred fibril structure, and thus its potential to induce aggregation of other variants. Overall, our results indicate that phosphorylation could play a key role in controlling aggregation propensity and may lead to the formation of distinct, non-cross-seeding fibril populations.
Collapse
Affiliation(s)
- Kalyani Sanagavarapu
- Biochemistry and Structural Biology, Department of Chemistry, Lund University Lund Sweden
| | - Georg Meisl
- Yusuf Hamied Chemistry Department, University of Cambridge Lensfield Road Cambridge UK
| | - Veronica Lattanzi
- Biochemistry and Structural Biology, Department of Chemistry, Lund University Lund Sweden
- Physical Chemistry, Department of Chemistry, Lund University Lund Sweden
| | - Katja Bernfur
- Biochemistry and Structural Biology, Department of Chemistry, Lund University Lund Sweden
| | - Birgitta Frohm
- Biochemistry and Structural Biology, Department of Chemistry, Lund University Lund Sweden
| | - Ulf Olsson
- Physical Chemistry, Department of Chemistry, Lund University Lund Sweden
| | - Tuomas P J Knowles
- Yusuf Hamied Chemistry Department, University of Cambridge Lensfield Road Cambridge UK
- Cavendish Laboratory, Department of Physics, University of Cambridge JJ Thomson Avenue Cambridge UK
| | - Anders Malmendal
- Biochemistry and Structural Biology, Department of Chemistry, Lund University Lund Sweden
- Department of Science and Environment, Roskilde University Roskilde Denmark
| | - Sara Linse
- Biochemistry and Structural Biology, Department of Chemistry, Lund University Lund Sweden
| |
Collapse
|
45
|
Pillai M, Jha SK. Conformational Enigma of TDP-43 Misfolding in Neurodegenerative Disorders. ACS OMEGA 2024; 9:40286-40297. [PMID: 39372031 PMCID: PMC11447851 DOI: 10.1021/acsomega.4c04119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/25/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024]
Abstract
Misfolding and aggregation of the protein remain some of the most common phenomena observed in neurodegeneration. While there exist multiple neurodegenerative disorders characterized by accumulation of distinct proteins, what remains particularly interesting is the ability of these proteins to undergo a conformational change to form aggregates. TDP-43 is one such nucleic acid binding protein whose misfolding is associated with many neurogenerative diseases including amyotrophic lateral sclerosis (ALS) and fronto-temporal lobar degeneration (FTLD). TDP-43 protein assumes several different conformations and oligomeric states under the diseased condition. In this review, we explore the intrinsic relationship between the conformational variability of TDP-43 protein, with a particular focus on the RRM domains, and its propensity to undergo aggregation. We further emphasize the probable mechanism behind the formation of these conformations and suggest a potential diagnostic and therapeutic strategy in the context of these conformational states of the protein.
Collapse
Affiliation(s)
- Meenakshi Pillai
- Physical
and Materials Chemistry Division, CSIR-National
Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Santosh Kumar Jha
- Physical
and Materials Chemistry Division, CSIR-National
Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
46
|
Mahdavimehr M, Kaboudin B, Alaie S, Tondkar F, Eshkaftaki ZM, Ebrahim-Habibi MB, Ghashghaee M, Tahmasebi E, Zhang T, Gu Y, Meratan AA. Inhibition of cytotoxic self-assembly of HEWL through promoting fibrillation by new synthesized α-hydroxycarbamoylphosphinic acids. RSC Adv 2024; 14:31227-31242. [PMID: 39355328 PMCID: PMC11443501 DOI: 10.1039/d4ra02969k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/04/2024] [Indexed: 10/03/2024] Open
Abstract
The main objective of the present study is to investigate the potency of new synthesized hydroxycarbamoyl phosphinic acid derivatives in modulating cytotoxic fibrillogenesis of hen egg white lysozyme (HEWL), as a common model in protein aggregation studies. Hydroxycarbamoyl phosphinic acid derivatives were prepared by the reaction of α-hydroxyalkylphosphinic acids with isocyanates (or isothiocyanates) in the presence of trimethylsilyl chloride (TMSCl). The designed process involves the condensation reaction leading to formation of new C sp2-P bond formation. The synthesis and purity of novel designed compounds were confirmed by NMR, LC-MS, and HPLC techniques. A range of experiments, including thioflavin T (ThT) and 8-anilino-1-naphthalenesulfonic acid (ANS) fluorescence assays, Congo red binding measurement, atomic force microscopy imaging, MTT-based cell viability and hemolysis assays were employed to investigate anti-amyloidogenic effects of tested compounds. The obtained results demonstrate that these compounds are able to significantly modulate the self-assembly process of HEWL via shortening of nucleation phase leading to the acceleration of fibrillation and appearance of very large and thick fibrils with decreased surface hydrophobicity and cytotoxicity. Based on ANS binding data, we suggest that increased exposure of hydrophobic patches of oligomeric species is the possible mechanism by which tested compounds promote self-assembly process of HEWL. Fluorescence anisotropy and molecular docking studies indicate the interaction of both synthesized compounds with HEWL, and more specifically with residues that are situated in the highly aggregation-prone β-domain region of protein. This study unveils the potential of hydroxyalkylphosphinic acids as modulators of amyloid fibrillation highlighting these compounds as a promising approach for targeting protein aggregates associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Mohsen Mahdavimehr
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Babak Kaboudin
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Saied Alaie
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Farimah Tondkar
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Zahra Mahmoudi Eshkaftaki
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | | | - Mojtaba Ghashghaee
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Elham Tahmasebi
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| | - Tianjian Zhang
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology Wuhan 430074 China
| | - Yanlong Gu
- School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology Wuhan 430074 China
| | - Ali Akbar Meratan
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS) Zanjan 45137-66731 Iran
| |
Collapse
|
47
|
Zhaliazka K, Kurouski D. Nanoscale Structural Characterization of Amyloid β 1-42 Oligomers and Fibrils Grown in the Presence of Fatty Acids. ACS Chem Neurosci 2024; 15:3344-3353. [PMID: 39222387 PMCID: PMC11413849 DOI: 10.1021/acschemneuro.4c00275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Mono- and polyunsaturated fatty acids (FAs) are broadly used as food supplements. However, their effect on the aggregation of amyloidogenic proteins remains unclear. In this study, we investigated the effect of a large number of mono- and polyunsaturated, as well as fully saturated FAs on the aggregation of amyloid β1-42 (Aβ1-42) peptide. A progressive aggregation of this peptide is the expected molecular cause of Alzheimer's disease (AD), one of the most common neurodegenerative pathologies in the world. We found that arachidonic and stearic acids delayed the aggregation of Aβ1-42. Using Nano-Infrared spectroscopy, we found that FAs caused very little if any changes in the secondary structure of Aβ1-42 oligomers and fibrils formed at different stages of protein aggregation. However, the analyzed mono- and polyunsaturated, as well as fully saturated FAs uniquely altered the toxicity of Aβ1-42 fibrils. We found a direct relationship between the degree of FAs unsaturation and toxicity of Aβ1-42 fibrils formed in their presence. Specifically, with an increase in the degree of unsaturation, the toxicity Aβ1-42/FA fibrils increased. These results indicate that fully saturated or monounsaturated FAs could be used to decrease the toxicity of amyloid aggregates and, consequently, decelerate the development of AD.
Collapse
Affiliation(s)
- Kiryl Zhaliazka
- Department
of Biochemistry and Biophysics, Texas A&M
University, College
Station, Texas 77843, United States
| | - Dmitry Kurouski
- Department
of Biochemistry and Biophysics, Texas A&M
University, College
Station, Texas 77843, United States
- Department
of Biomedical Engineering, Texas A&M
University, College Station, Texas 77843, United States
| |
Collapse
|
48
|
Dabas A, Goyal B. Structural Reorganization Mechanism of the Aβ 42 Fibril Mediated by N-Substituted Oligopyrrolamide ADH-353. ACS Chem Neurosci 2024; 15:3136-3151. [PMID: 39158263 DOI: 10.1021/acschemneuro.4c00253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024] Open
Abstract
The inhibition of amyloid-β (Aβ) fibrillation and clearance of Aβ aggregates have emerged as a potential pharmacological strategy to alleviate Aβ aggregate-induced neurotoxicity in Alzheimer's disease (AD). Maity et al. shortlisted ADH-353 from a small library of positively charged N-substituted oligopyrrolamides for its notable ability to inhibit Aβ fibrillation, disintegrate intracellular cytotoxic Aβ oligomers, and alleviate Aβ-induced cytotoxicity in the SH-SY5Y and N2a cells. However, the molecular mechanism through which ADH-353 interacts with the Aβ42 fibrils, leading to their disruption and subsequent clearance, remains unclear. Thus, a detailed molecular mechanism underlying the disruption of neurotoxic Aβ42 fibrils (PDB ID 2NAO) by ADH-353 has been illuminated in this work using molecular dynamics simulations. Interestingly, conformational snapshots during simulation depicted the shortening and disappearance of β-strands and the emergence of a helix conformation, indicating a loss of the well-organized β-sheet-rich structure of the disease-relevant Aβ42 fibril on the incorporation of ADH-353. ADH-353 binds strongly to the Aβ42 fibril (ΔGbinding= -142.91 ± 1.61 kcal/mol) with a notable contribution from the electrostatic interactions between positively charged N-propylamine side chains of ADH-353 with the glutamic (Glu3, Glu11, and Glu22) and aspartic (Asp7 and Asp23) acid residues of the Aβ42 fibril. This aligns well with heteronuclear single quantum coherence NMR studies, which depict that the binding of ADH-353 with the Aβ peptide is driven by electrostatic and hydrophobic contacts. Furthermore, a noteworthy decrease in the binding affinity of Aβ42 fibril chains on the incorporation of ADH-353 indicates the weakening of interchain interactions leading to the disruption of the double-horseshoe conformation of the Aβ42 fibril. The illumination of key interactions responsible for the destabilization of the Aβ42 fibril by ADH-353 in this work will greatly aid in designing new chemical scaffolds with enhanced efficacy for the clearance of Aβ aggregates in AD.
Collapse
Affiliation(s)
- Arushi Dabas
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| | - Bhupesh Goyal
- Department of Chemistry & Biochemistry, Thapar Institute of Engineering & Technology, Patiala, Punjab 147004, India
| |
Collapse
|
49
|
Chiang W, Urban JM, Yanchik-Slade F, Stout A, Hammond JM, Nilsson BL, Gelbard HA, Krauss TD. Hybrid Amyloid Quantum Dot Nano-Bio Assemblies to Probe Neuroinflammatory Damage. ACS Chem Neurosci 2024; 15:3124-3135. [PMID: 39146244 PMCID: PMC11378299 DOI: 10.1021/acschemneuro.4c00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/29/2024] [Accepted: 08/07/2024] [Indexed: 08/17/2024] Open
Abstract
Various oligomeric species of amyloid-beta have been proposed to play different immunogenic roles in the cellular pathology of Alzheimer's Disease. The dynamic interconversion between various amyloid oligomers and fibrillar assemblies makes it difficult to elucidate the role each potential aggregation state may play in driving neuroinflammatory and neurodegenerative pathology. The ability to identify the amyloid species that are key and essential drivers of these pathological hallmarks of Alzheimer's Disease is of fundamental importance for also understanding downstream events including tauopathies that mediate neuroinflammation with neurologic deficits. Here, we report the design and construction of a quantum dot mimetic for larger spherical oligomeric amyloid species as an "endogenously" fluorescent proxy for this cytotoxic assembly of amyloid to investigate its role in inducing inflammatory and stress response states in neuronal and glial cell types. The design parameters and construction protocol developed here may be adapted for developing quantum dot nano-bio assemblies for other biological systems of interest, particularly neurodegenerative diseases involving other protein aggregates.
Collapse
Affiliation(s)
- Wesley Chiang
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
- Department
of Biochemistry and Biophysics, University
of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jennifer M. Urban
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Francine Yanchik-Slade
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Angela Stout
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jennetta M. Hammond
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Bradley L. Nilsson
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Harris A. Gelbard
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
- Departments
of Pediatrics, Neuroscience, and Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Todd D. Krauss
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
- The
Institute of Optics, University of Rochester
Medical Center, Rochester, New York 14627-0216, United States
| |
Collapse
|
50
|
Palmioli A, Airoldi C. An NMR Toolkit to Probe Amyloid Oligomer Inhibition in Neurodegenerative Diseases: From Ligand Screening to Dissecting Binding Topology and Mechanisms of Action. Chempluschem 2024; 89:e202400243. [PMID: 38712695 DOI: 10.1002/cplu.202400243] [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: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 05/08/2024]
Abstract
The aggregation of amyloid peptides and proteins into toxic oligomers is a hallmark of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Machado-Joseph's disease, and transmissible spongiform encephalopathies. Inhibition of amyloid oligomers formation and interactions with biological counterparts, as well as the triggering of non-toxic amorphous aggregates, are strategies towards preventive interventions against these pathologies. NMR spectroscopy addresses the need for structural characterization of amyloid proteins and their aggregates, their binding to inhibitors, and rapid screening of compound libraries for ligand identification. Here we briefly discuss the solution experiments constituting the NMR spectroscopist's toolkit and provide examples of their application.
Collapse
Affiliation(s)
- Alessandro Palmioli
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126, Milan, Italy
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, University of Milano - Bicocca, P.zza della Scienza 2, 20126, Milan, Italy
| |
Collapse
|