Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biomed Res Int ; 2024: 9985719, 2024.
Article in English | MEDLINE | ID: mdl-38221912

ABSTRACT

Introduction: Alzheimer's disease (AD) is a neurodegenerative disorder with no conclusive remedy. Yohimbine, found in Rauwolfia vomitoria, may reduce brain inflammation by targeting tumour necrosis factor-alpha (TNFα), implicated in AD pathogenesis. Metoserpate, a synthetic compound, may inhibit TNFα. The study is aimed at assessing the potential utility of repurposing metoserpate for TNFα inhibition to reduce neuronal damage and inflammation in AD. The development of safe and effective treatments for AD is crucial to address the growing burden of the disease, which is projected to double over the next two decades. Methods: Our study repurposed an FDA-approved drug as TNFα inhibitor for AD management using structural similarity studies, molecular docking, and molecular dynamics simulations. Yohimbine was used as a reference compound. Molecular docking used SeeSAR, and molecular dynamics simulation used GROMACS. Results: Metoserpate was selected from 10 compounds similar to yohimbine based on pharmacokinetic properties and FDA approval status. Molecular docking and simulation studies showed a stable interaction between metoserpate and TNFα over 100 ns (100000 ps). This suggests a reliable and robust interaction between the protein and ligand, supporting the potential utility of repurposing metoserpate for TNFα inhibition in AD treatment. Conclusion: Our study has identified metoserpate, a previously FDA-approved antihypertensive agent, as a promising candidate for inhibiting TNFα in the management of AD.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Molecular Docking Simulation , Tumor Necrosis Factor-alpha , Drug Repositioning , Molecular Dynamics Simulation , Yohimbine/pharmacology , Yohimbine/therapeutic use
2.
Front Neurosci ; 17: 1132670, 2023.
Article in English | MEDLINE | ID: mdl-37034163

ABSTRACT

Oxidative stress is a significant source of damage that accumulates during aging and contributes to Alzheimer's disease (AD) pathogenesis. Oxidation of proteins can give rise to covalent links between adjacent tyrosines known as dityrosine (DiY) cross-linking, amongst other modifications, and this observation suggests that DiY could serve as a biomarker of accumulated oxidative stress over the lifespan. Many studies have focused on understanding the contribution of DiY to AD pathogenesis and have revealed that DiY crosslinks can be found in both Aß and tau deposits - the two key proteins involved in the formation of amyloid plaques and tau tangles, respectively. However, there is no consensus yet in the field on the impact of DiY on Aß and tau function, aggregation, and toxicity. Here we review the current understanding of the role of DiY on Aß and tau gathered over the last 20 years since the first observation, and discuss the effect of this modification for Aß and tau aggregation, and its potential as a biomarker for AD.

3.
Essays Biochem ; 66(7): 1001-1011, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36373666

ABSTRACT

Tau is an intrinsically disordered protein that has the ability to self-assemble to form paired helical and straight filaments in Alzheimer's disease, as well as the ability to form additional distinct tau filaments in other tauopathies. In the presence of microtubules, tau forms an elongated form associated with tubulin dimers via a series of imperfect repeats known as the microtubule binding repeats. Tau has recently been identified to have the ability to phase separate in vitro and in cells. The ability of tau to adopt a wide variety of conformations appears fundamental both to its biological function and also its association with neurodegenerative diseases. The recently highlighted involvement of low-complexity domains in liquid-liquid phase separation provides a critical link between the soluble function and the insoluble dysfunctional properties of tau.


Subject(s)
Alzheimer Disease , tau Proteins , Humans , tau Proteins/chemistry , tau Proteins/metabolism , Neurofibrillary Tangles/metabolism , Alzheimer Disease/metabolism , Microtubules/metabolism , Tubulin/metabolism
4.
J Mol Biol ; 434(19): 167785, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35961386

ABSTRACT

A characteristic hallmark of Alzheimer's Disease (AD) is the pathological aggregation and deposition of tau into paired helical filaments (PHF) in neurofibrillary tangles (NFTs). Oxidative stress is an early event during AD pathogenesis and is associated with tau-mediated AD pathology. Oxidative environments can result in the formation of covalent dityrosine crosslinks that can increase protein stability and insolubility. Dityrosine cross-linking has been shown in Aß plaques in AD and α-synuclein aggregates in Lewy bodies in ex vivo tissue sections, and this modification may increase the insolubility of these aggregates and their resistance to degradation. Using the PHF-core tau fragment (residues 297 - 391) as a model, we have previously demonstrated that dityrosine formation traps tau assemblies to reduce further elongation. However, it is unknown whether dityrosine crosslinks are found in tau deposits in vivo in AD and its relevance to disease mechanism is unclear. Here, using transmission electron microscope (TEM) double immunogold-labelling, we reveal that neurofibrillary NFTs in AD are heavily decorated with dityrosine crosslinks alongside tau. Single immunogold-labelling TEM and fluorescence spectroscopy revealed the presence of dityrosine on AD brain-derived tau oligomers and fibrils. Using the tau (297-391) PHF-core fragment as a model, we further showed that prefibrillar tau species are more amenable to dityrosine crosslinking than tau fibrils. Dityrosine formation results in heat and SDS stability of oxidised prefibrillar and fibrillar tau assemblies. This finding has implications for understanding the mechanism governing the insolubility and toxicity of tau assemblies in vivo.


Subject(s)
Alzheimer Disease , Neurofibrillary Tangles , Tyrosine , tau Proteins , Alzheimer Disease/metabolism , Humans , Neurofibrillary Tangles/chemistry , Protein Conformation, alpha-Helical , Tyrosine/analogs & derivatives , Tyrosine/chemistry , alpha-Synuclein/chemistry , tau Proteins/chemistry
5.
Cells ; 10(3)2021 03 22.
Article in English | MEDLINE | ID: mdl-33809978

ABSTRACT

The self-assembly of tau into paired helical filaments (PHFs) in neurofibrillary tangles (NFTs) is a significant event in Alzheimer's disease (AD) pathogenesis. Numerous post-translational modifications enhance or inhibit tau assembly into NFTs. Oxidative stress, which accompanies AD, induces multiple post-translational modifications in proteins, including the formation of dityrosine (DiY) cross-links. Previous studies have revealed that metal-catalysed oxidation (MCO) using Cu2+ and H2O2 leads to the formation of DiY cross-links in two misfolding proteins, Aß and α-synuclein, associated with AD and Parkinson's disease respectively. The effect of MCO on tau remains unknown. Here, we examined the effect of MCO and ultra-violet oxidation to study the influence of DiY cross-linking on the self-assembly of the PHF-core tau fragment. We report that DiY cross-linking facilitates tau assembly into tau oligomers that fail to bind thioflavin S, lack ß-sheet structure and prevents their elongation into filaments. At a higher concentration, Cu2+ (without H2O2) also facilitates the formation of these tau oligomers. The DiY cross-linked tau oligomers do not cause cell death. Our findings suggest that DiY cross-linking of pre-assembled tau promotes the formation of soluble tau oligomers that show no acute impact on cell viability.


Subject(s)
Neurons/metabolism , Oxidative Stress , Peptide Fragments/metabolism , Protein Processing, Post-Translational , Reactive Oxygen Species/metabolism , tau Proteins/metabolism , Cell Line, Tumor , Chelating Agents/pharmacology , Copper/pharmacology , Edetic Acid/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Neurons/drug effects , Neurons/pathology , Oxidants/pharmacology , Oxidative Stress/drug effects , Protein Conformation, beta-Strand , Protein Multimerization , Protein Processing, Post-Translational/drug effects , Solubility , Structure-Activity Relationship
6.
J Mol Biol ; 433(3): 166732, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33279578

ABSTRACT

An N-terminal hepta-peptide sequence of yeast prion protein Sup35 with the sequence GNNQQNY is widely used as a model system for amyloid fibril formation. In this study, we used a reproducible solubilisation protocol that allows the generation of a homogenous monomeric solution of GNNQQNY to uncover the molecular details of its self-assembly mechanism. The aggregation kinetics data show that the GNNQQNY sequence follows nucleation-dependent aggregation kinetics with a critical nucleus of size ~7 monomers and that the efficiency of nucleation were found to be inversely related to the reaction temperature. The nucleus reduces the thermodynamic energy barrier by acting as a template for further self-assembly and results in highly ordered amyloid fibrils. The fibers grown at different temperatures showed similar Thioflavin T fluorescence, Congo-red binding and ß-sheet rich structures displaying a characteristic cross-ß diffraction pattern. These aggregates also share morphological and structural identity with those reported earlier. The mature GNNQQNY fibers did not exert significant oxidative stress or cytotoxicity upon incubating with differentiated SHSY5Y cells. To our knowledge, this is the first study to experimentally validate previous nucleus size predictions based on theoretical and molecular dynamics simulations. These findings provide the basis for understanding the kinetics and thermodynamics of amyloid nucleation and elongation of amyloidogenic proteins/peptides associated with many systemic and neurodegenerative diseases.


Subject(s)
Amino Acid Sequence , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Peptide Fragments/chemistry , Protein Aggregates , Yeasts , Amyloid/metabolism , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/metabolism , Cell Survival , Fluorescent Antibody Technique , Kinetics , Peptide Fragments/metabolism , Protein Transport , Solubility , Spectrum Analysis , Thermodynamics , Yeasts/metabolism
7.
Front Neurol ; 11: 590754, 2020.
Article in English | MEDLINE | ID: mdl-33281730

ABSTRACT

Tau plays an important pathological role in a group of neurodegenerative diseases called tauopathies, including Alzheimer's disease, Pick's disease, chronic traumatic encephalopathy and corticobasal degeneration. In each disease, tau self-assembles abnormally to form filaments that deposit in the brain. Tau is a natively unfolded protein that can adopt distinct structures in different pathological disorders. Cryo-electron microscopy has recently provided a series of structures for the core of the filaments purified from brain tissue from patients with different tauopathies and revealed that they share a common core region, while differing in their specific conformation. This structurally resolvable part of the core is contained within a proteolytically stable core region from the repeat domain initially isolated from AD tau filaments. Tau has recently become an important target for therapy. Recent work has suggested that the prevention of tau self-assembly may be effective in slowing the progression of Alzheimer's disease and other tauopathies. Here we review the work that explores the importance of tau filament structures and tau self-assembly mechanisms, as well as examining model systems that permit the exploration of the mode of action of potential inhibitors.

8.
iScience ; 23(10): 101537, 2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33083713

ABSTRACT

Dityrosine (DiY), via the cross-linking of tyrosine residues, is a marker of protein oxidation, which increases with aging. Amyloid-ß (Aß) forms DiY in vitro and DiY-cross-linked Aß is found in the brains of patients with Alzheimer disease. Metal- or UV- catalyzed oxidation of Aß42 results in an increase in DiY cross-links. Using DiY as a marker of oxidation, we compare the self-assembly propensity and DiY cross-link formation for a non-assembly competent variant of Aß42 (vAß) with wild-type Aß42. Oxidation results in the formation of trapped wild-type Aß assemblies with increased DiY cross-links that are unable to elongate further. Assembly-incompetent vAß and trapped Aß assemblies are non-toxic to neuroblastoma cells at all stages of self-assembly, in contrast to oligomeric, non-cross-linked Aß. These findings point to a mechanism of toxicity that necessitates dynamic self-assembly whereby trapped Aß assemblies and assembly-incompetent variant Aß are unable to result in cell death.

9.
Front Cell Neurosci ; 12: 220, 2018.
Article in English | MEDLINE | ID: mdl-30123109

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia and is distinguished from other dementias by observation of extracellular Amyloid-ß (Aß) plaques and intracellular neurofibrillary tangles, comprised of fibrils of Aß and tau protein, respectively. At early stages, AD is characterized by minimal neurodegeneration, oxidative stress, nucleolar stress, and altered protein synthesis machinery. It is generally believed that Aß oligomers are the neurotoxic species and their levels in the AD brain correlate with the severity of dementia suggesting that they play a critical role in the pathogenesis of the disease. Here, we show that the incubation of differentiated human neuroblastoma cells (SHSY5Y) with freshly prepared Aß42 oligomers initially resulted in oxidative stress and subtle nucleolar stress in the absence of DNA damage or cell death. The presence of exogenous Aß oligomers resulted in altered nuclear tau levels as well as phosphorylation state, leading to altered distribution of nucleolar tau associated with nucleolar stress. These markers of cellular dysfunction worsen over time alongside a reduction in ribosomal RNA synthesis and processing, a decrease in global level of newly synthesized RNA and reduced protein synthesis. The interplay between Aß and tau in AD remains intriguing and Aß toxicity has been linked to tau phosphorylation and changes in localization. These findings provide evidence for the involvement of Aß42 effects on nucleolar tau and protein synthesis machinery dysfunction in cultured cells. Protein synthesis dysfunction is observed in mild cognitive impairment and early AD in the absence of significant neuronal death.

10.
Acta Neuropathol Commun ; 6(1): 70, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30064522

ABSTRACT

Tau is known for its pathological role in neurodegenerative diseases, including Alzheimer's disease (AD) and other tauopathies. Tau is found in many subcellular compartments such as the cytosol and the nucleus. Although its normal role in microtubule binding is well established, its nuclear role is still unclear. Here, we reveal that tau localises to the nucleolus in undifferentiated and differentiated neuroblastoma cells (SHSY5Y), where it associates with TIP5, a key player in heterochromatin stability and ribosomal DNA (rDNA) transcriptional repression. Immunogold labelling on human brain sample confirms the physiological relevance of this finding by showing tau within the nucleolus colocalises with TIP5. Depletion of tau results in an increase in rDNA transcription with an associated decrease in heterochromatin and DNA methylation, suggesting that under normal conditions tau is involved in silencing of the rDNA. Cellular stress induced by glutamate causes nucleolar stress associated with the redistribution of nucleolar non-phosphorylated tau, in a similar manner to fibrillarin, and nuclear upsurge of phosphorylated tau (Thr231) which doesn't colocalise with fibrillarin or nucleolar tau. This suggests that stress may impact on different nuclear tau species. In addition to involvement in rDNA transcription, nucleolar non-phosphorylated tau also undergoes stress-induced redistribution similar to many nucleolar proteins.


Subject(s)
Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Excitatory Amino Acid Agonists/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glutamic Acid/pharmacology , tau Proteins/metabolism , Brain/metabolism , Brain/ultrastructure , Cell Differentiation/physiology , Cell Line, Tumor , Cell Nucleolus/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/ultrastructure , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Gene Expression Regulation, Neoplastic/genetics , Heterochromatin/physiology , Histones/metabolism , Humans , Immunoprecipitation , Microscopy, Confocal , Microscopy, Electron , Neuroblastoma/pathology , Neuroblastoma/ultrastructure , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Transport/drug effects , RNA, Messenger , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic/drug effects , Transfection , tau Proteins/genetics , tau Proteins/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...