Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Commun Signal ; 22(1): 233, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38641599

RESUMEN

BACKGROUND: Multiple neurodegenerative diseases are induced by the formation and deposition of protein aggregates. In particular, the microtubule-associated protein Tau leads to the development of so-called tauopathies characterized by the aggregation of hyperphosphorylated Tau within neurons. We recently showed that the constitutive activity of the serotonin receptor 7 (5-HT7R) is required for Tau hyperphosphorylation and aggregation through activation of the cyclin-dependent kinase 5 (CDK5). We also demonstrated physical interaction between 5-HT7R and CDK5 at the plasma membrane suggesting that the 5-HT7R/CDK5 complex is an integral part of the signaling network involved in Tau-mediated pathology. METHODS: Using biochemical, microscopic, molecular biological, computational and AI-based approaches, we investigated structural requirements for the formation of 5-HT7R/CDK5 complex. RESULTS: We demonstrated that 5-HT7R domains responsible for coupling to Gs proteins are not involved in receptor interaction with CDK5. We also created a structural model of the 5-HT7R/CDK5 complex and refined the interaction interface. The model predicted two conserved phenylalanine residues, F278 and F281, within the third intracellular loop of 5-HT7R to be potentially important for complex formation. While site-directed mutagenesis of these residues did not influence Gs protein-mediated receptor signaling, replacement of both phenylalanines by alanine residues significantly reduced 5-HT7R/CDK5 interaction and receptor-mediated CDK5 activation, leading to reduced Tau hyperphosphorylation and aggregation. Molecular dynamics simulations of 5-HT7R/CDK5 complex for wild-type and receptor mutants confirmed binding interface stability of the initial model. CONCLUSIONS: Our results provide a structural basis for the development of novel drugs targeting the 5-HT7R/CDK5 interaction interface for the selective treatment of Tau-related disorders, including frontotemporal dementia and Alzheimer's disease.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina , Activación Enzimática , Receptores de Serotonina , Humanos , Enfermedad de Alzheimer/metabolismo , Quinasa 5 Dependiente de la Ciclina/química , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Fosforilación , Receptores de Serotonina/química , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Transducción de Señal
2.
PLoS One ; 19(4): e0297903, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38626021

RESUMEN

Social networks often involve the users rating each other based on their beliefs, abilities, and other characteristics. This is particularly common in e-commerce platforms where buyers rate sellers based on their trustworthiness. However, the rating tends to vary between users due to differences in their individual scoring criteria. For example, in a transaction network, a positive user may give a high rating unless the transaction was unsatisfactory while a neutral user may give a mid-rating, consequently giving the same numeric score to different levels of satisfaction. In this paper, we propose a novel method called user tendency-based rating scaling, which adjusts the current rating (its score) based on the pattern of past ratings. We investigate whether this rating scaling method can classify between "good users" and "bad users" in online trade social networks better when compared with using the original rating scores without scaling. Classifying between good users and bad users is especially important for anonymous rating networks like Bitcoin transaction networks, where users' reputations must be recorded to preclude fraudulent and risky users. We evaluate the proposed rating scaling method by performing user classification, link prediction, and clustering tasks, using three real-world online rating network datasets. We use both the original ratings and the scaled ratings as weights of graphs and use a weighted graph embedding method to find node representations that reflect users' positive and negative information. The experimental results showed that using the proposed rating scaling method outperformed using the original (i.e., unscaled) ratings by up to 17% in classification accuracy, and by up to 2.5% in link prediction based on the AUC ROC measure, and by up to 21% in the clustering tasks based on the Dunn-index.


Asunto(s)
Comercio , Red Social
3.
Angew Chem Int Ed Engl ; 63(3): e202312942, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38062619

RESUMEN

The development of a small-molecule probe designed to selectively target neurons would enhance the exploration of intricate neuronal structures and functions. Among such probes, NeuO stands out as the pioneer and has gained significant traction in the field of research. Nevertheless, neither the mechanism behind neuron-selectivity nor the cellular localization has been determined. Here, we introduce NeuM, a derivative of NeuO, designed to target neuronal cell membranes. Furthermore, we elucidate the mechanism behind the selective neuronal membrane trafficking that distinguishes neurons. In an aqueous buffer, NeuM autonomously assembles into micellar structures, leading to the quenching of its fluorescence (Φ=0.001). Upon exposure to neurons, NeuM micelles were selectively internalized into neuronal endosomes via clathrin-mediated endocytosis. Through the endocytic recycling pathway, NeuM micelles integrate into neuronal membrane, dispersing fluorescent NeuM molecules in the membrane (Φ=0.61). Molecular dynamics simulations demonstrated that NeuM, in comparison to NeuO, possesses optimal lipophilicity and molecular length, facilitating its stable incorporation into phospholipid layers. The stable integration of NeuM within neuronal membrane allows the prolonged monitoring of neurons, as well as the visualization of intricate neuronal structures.


Asunto(s)
Clatrina , Micelas , Clatrina/metabolismo , Endocitosis/fisiología , Endosomas/metabolismo , Neuronas/metabolismo
4.
Chemistry ; 30(5): e202302916, 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-37902438

RESUMEN

Blood continually contributes to the maintenance of homeostasis of the body and contains information regarding the health state of an individual. However, current hematological analyses predominantly rely on a limited number of CD markers and morphological analysis. In this work, differentially sensitive fluorescent compounds based on TCF scaffolds are introduced that are designed for fluorescent phenotyping of blood. Depending on their structures, TCF compounds displayed varied responses to reactive oxygen species, biothiols, redox-related biomolecules, and hemoglobin, which are the primary influential factors within blood. Contrary to conventional CD marker-based analysis, this unbiased fluorescent phenotyping method produces diverse fingerprints of the health state. Precise discrimination of blood samples from 37 mice was demonstrated based on their developmental stages, ranging from 10 to 19 weeks of age. Additionally, this fluorescent phenotyping method enabled the differentiation between drugs with distinct targets, serving as a simple yet potent tool for pharmacological analysis to understand the mode of action of various drugs.


Asunto(s)
Envejecimiento , Colorantes Fluorescentes , Ratones , Animales , Colorantes Fluorescentes/química , Especies Reactivas de Oxígeno/análisis , Oxidación-Reducción , Células Sanguíneas/química
5.
Bioorg Med Chem ; 95: 117513, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37931520

RESUMEN

In this article, the development of fluorescent imaging probes for the detection of Alzheimer's disease (AD)-associated protein aggregates is described. Indane derivatives with a donor-π-acceptor (D-π-A) structure were designed and synthesized. The probes were evaluated for their ability to bind to ß-amyloid (Aß) protein aggregates, which are a key pathological hallmark of AD. The results showed that several probes exhibited significant changes in fluorescence intensity at wavelengths greater than 600 nm when they were bound to Aß aggregates compared to the Aß monomeric form. Among the tested probes, four D-π-A type indane derivatives showed promising binding selectivity to Aß aggregates over non-specific proteins such as bovine serum albumin (BSA). The molecular docking study showed that our compounds were appropriately located along the Aß fibril axis through the hydrophobic tunnel structure. Further analysis revealed that the most active compound having dimethylaminopyridyl group as an election donor and dicyano group as an electron acceptor could effectively stain Aß plaques in brain tissue samples from AD transgenic mice. These findings suggest that our indane-based compounds have the potential to serve as fluorescent probes for the detection and monitoring of Aß aggregation in AD.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Colorantes Fluorescentes/química , Agregado de Proteínas , Simulación del Acoplamiento Molecular , Péptidos beta-Amiloides/metabolismo , Ratones Transgénicos , Encéfalo/metabolismo , Placa Amiloide/química , Placa Amiloide/diagnóstico , Placa Amiloide/patología
6.
Nanoscale ; 15(21): 9315-9328, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37158478

RESUMEN

Amyloid generation plays essential roles in various human diseases, biological functions, and nanotechnology. However, developing efficient chemical and biological candidates for regulating amyloid fibrillation remains difficult because information on the molecular actions of modulators is insufficient. Thus, studies are needed to understand how the intermolecular physicochemical properties of the synthesised molecules and amyloid precursors influence amyloidogenesis. In this study, we synthesised a novel amphiphilic sub-nanosized material, arginine-arginine (RR)-bile acid (BA), by conjugating positively charged RR to hydrophobic BA. The effects of RR-BA on amyloid formation were investigated on α-synuclein (αSN) in Parkinson's disease and on K18 and amyloid-ß (1-42) (Aß42) in Alzheimer's disease. RR-BA showed no appreciable effect on the kinetics of K18 and Aß42 amyloid fibrillation because of their weak and non-specific interactions. However, RR-BA specifically bound to αSN with moderate binding affinity through electrostatic interactions between the positively charged RR and the negatively charged cluster in the C-terminus of αSN. In addition, hydrophobic BA in the αSN-RR-BA complex transiently condensed αSN for primary nucleation, thereby accelerating αSN amyloid fibrillation. We propose an electrostatic binding and hydrophobic condensation model of RR-BA-driven amyloid formation of αSN, which will contribute to the rational design and development of molecules for controlling amyloid aggregation in diverse fields.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Parkinson , Humanos , alfa-Sinucleína/química , Enfermedad de Parkinson/metabolismo , Amiloide/química , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides
7.
Alzheimers Dement ; 19(12): 5482-5497, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37218673

RESUMEN

INTRODUCTION: Hyperphosphorylation and aggregation of the microtubule-associated protein tau cause the development of tauopathies, such as Alzheimer's disease and frontotemporal dementia (FTD). We recently uncovered a causal link between constitutive serotonin receptor 7 (5-HT7R) activity and pathological tau aggregation. Here, we evaluated 5-HT7R inverse agonists as novel drugs in the treatment of tauopathies. METHODS: Based on structural homology, we screened multiple approved drugs for their inverse agonism toward 5-HT7R. Therapeutic potential was validated using biochemical, pharmacological, microscopic, and behavioral approaches in different cellular models including tau aggregation cell line HEK293 tau bimolecular fluorescence complementation, primary mouse neurons, and human induced pluripotent stem cell-derived neurons carrying an FTD-associated tau mutation as well as in two mouse models of tauopathy. RESULTS: Antipsychotic drug amisulpride is a potent 5-HT7R inverse agonist. Amisulpride ameliorated tau hyperphosphorylation and aggregation in vitro. It further reduced tau pathology and abrogated memory impairment in mice. DISCUSSION: Amisulpride may be a disease-modifying drug for tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Demencia Frontotemporal , Células Madre Pluripotentes Inducidas , Tauopatías , Humanos , Ratones , Animales , Agonismo Inverso de Drogas , Amisulprida/uso terapéutico , Demencia Frontotemporal/tratamiento farmacológico , Demencia Frontotemporal/genética , Células HEK293 , Células Madre Pluripotentes Inducidas/metabolismo , Tauopatías/genética , Proteínas tau/metabolismo , Enfermedad de Alzheimer/patología
8.
Exp Mol Med ; 55(3): 612-627, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36914856

RESUMEN

Tau oligomers play critical roles in tau pathology and are responsible for neuronal cell death and transmitting the disease in the brain. Accordingly, preventing tau oligomerization has become an important therapeutic strategy to treat tauopathies, including Alzheimer's disease. However, progress has been slow because detecting tau oligomers in the cellular context is difficult. Working toward tau-targeted drug discovery, our group has developed a tau-BiFC platform to monitor and quantify tau oligomerization. By using the tau-BiFC platform, we screened libraries with FDA-approved and passed phase I drugs and identified levosimendan as a potent anti-tau agent that inhibits tau oligomerization. 14C-isotope labeling of levosimendan revealed that levosimendan covalently bound to tau cysteines, directly inhibiting disulfide-linked tau oligomerization. In addition, levosimendan disassembles tau oligomers into monomers, rescuing neurons from aggregation states. In comparison, the well-known anti-tau agents methylene blue and LMTM failed to protect neurons from tau-mediated toxicity, generating high-molecular-weight tau oligomers. Levosimendan displayed robust potency against tau oligomerization and rescued cognitive declines induced by tauopathy in the TauP301L-BiFC mouse model. Our data present the potential of levosimendan as a disease-modifying drug for tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Tauopatías , Ratones , Animales , Proteínas tau/metabolismo , Simendán/farmacología , Simendán/uso terapéutico , Simendán/metabolismo , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo , Tauopatías/patología , Enfermedad de Alzheimer/metabolismo , Neuronas/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos
9.
Biomedicines ; 10(12)2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36551947

RESUMEN

Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide, causing progressive cognitive decline, memory impairment, and neurological deficits. Methylene blue (MB), an antioxidant, has emerged as a potential drug for the treatment of AD owing to its cognitive improvement and neuroprotective functions. Despite the small molecular size of MB, which can cross the BBB, the therapeutic effective dosage using a BBB-permeable delivery system in a specific brain localization remains unclear. In this study, we presented magnetic resonance-guided focused ultrasound (MRgFUS) as a delivery system to enhance BBB permeability for the effective treatment of AD. MRgFUS using two ultrasound intensities (0.25 and 0.32 MPa) was used to intravenously deliver MB to the hippocampal region. Compared with treatment with 0.25 MPa FUS, treatment with 0.32 MPa FUS significantly enhanced MB brain accumulation. Deposition of amyloid-ß (Aß) plaques and neural cell damage was significantly reduced in 0.32 MPa FUS/MB-treated APP/PS1 mice. Furthermore, aquaporin-4 expression increased significantly in the 0.32 MPa FUS and 0.32 MPa FUS/MB groups without glial fibrillary acidic protein activation. The results from this study demonstrate that FUS improved MB delivery to the brain, and FUS/MB combination treatment reduced the number of Aß plaques. This study revealed the potential of FUS-BBBD as an effective strategy to enhance the efficacy of therapeutic drugs for AD.

10.
Front Aging Neurosci ; 14: 932541, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35923541

RESUMEN

Neuronal accumulation of mis-folded tau is the pathological hallmark of multiple neurodegenerative disorders, including Alzheimer's disease. Distinct from amyloid plaques, which appear simultaneously throughout the brain, tau pathology develops first in a specific brain region and then propagates to neuroanatomically connected brain regions, exacerbating the disease. Due to the implication in disease progression, prevention of tau transmission is recognized as an important therapeutic strategy that can halt disease progression in the brain. Recently, accumulating studies have demonstrated diverse cellular mechanisms associated with cell-to-cell transmission of tau. Once transmitted, mis-folded tau species act as a prion-like seed for native tau aggregation in the recipient neuron. In this review, we summarize the diverse cellular mechanisms associated with the secretion and uptake of tau, and highlight tau-trafficking receptors, which mediate tau clearance or cell-to-cell tau transmission.

11.
Artículo en Inglés | MEDLINE | ID: mdl-35613066

RESUMEN

In recent years, many recommender systems using network embedding (NE) such as graph neural networks (GNNs) have been extensively studied in the sense of improving recommendation accuracy. However, such attempts have focused mostly on utilizing only the information of positive user-item interactions with high ratings. Thus, there is a challenge on how to make use of low rating scores for representing users' preferences since low ratings can be still informative in designing NE-based recommender systems. In this study, we present SiReN, a new Sign-aware Recommender system based on GNN models. Specifically, SiReN has three key components: 1) constructing a signed bipartite graph for more precisely representing users' preferences, which is split into two edge-disjoint graphs with positive and negative edges each; 2) generating two embeddings for the partitioned graphs with positive and negative edges via a GNN model and a multilayer perceptron (MLP), respectively, and then using an attention model to obtain the final embeddings; and 3) establishing a sign-aware Bayesian personalized ranking (BPR) loss function in the process of optimization. Through comprehensive experiments, we empirically demonstrate that SiReN consistently outperforms state-of-the-art NE-aided recommendation methods.

13.
J Am Chem Soc ; 144(11): 5067-5073, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35258954

RESUMEN

Aggregation of amyloidogenic proteins causing neurodegenerative diseases is an uncontrollable and contagious process that is often associated with lipid membranes in a highly complex physiological environment. Although several approaches using natural cells and membrane models have been reported, systematic investigations focusing on the association with the membranes are highly challenging, mostly because of the lack of proper molecular tools. Here, we report a new supramolecular approach using a synthetic cell system capable of controlling the initiation of protein aggregation and mimicking various conditions of lipid membranes, thereby enabling systematic investigations of membrane-dependent effects on protein aggregation by visualization. Extending this strategy through concurrent use of synthetic cells and natural cells, we demonstrate the potential of this approach for systematic and in-depth studies on interrogating inter- and intracellularly transmittable protein aggregation. Thus, this new approach offers opportunities for gaining insights into the pathological implications of contagious protein aggregation associated with membranes for neurotoxicity.


Asunto(s)
Células Artificiales , Proteínas Amiloidogénicas/metabolismo , Membrana Celular/metabolismo , Humanos , Lípidos , Agregado de Proteínas , Agregación Patológica de Proteínas
14.
Bioact Mater ; 13: 239-248, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35224305

RESUMEN

The pathological origin of Alzheimer's disease (AD) is still shrouded in mystery, despite intensive worldwide research efforts. The selective visualization of ß-amyloid (Aß), the most abundant proteinaceous deposit in AD, is pivotal to reveal AD pathology. To date, several small-molecule fluorophores for Aß species have been developed, with increasing binding affinities. In the current work, two organic small-molecule dioxaborine-derived fluorophores were rationally designed through tailoring the hydrophobicity with the aim to enhance the binding affinity for Aß1-42 fibrils -while concurrently preventing poor aqueous solubility-via biannulate donor motifs in D-π-A dyes. An unprecedented sub-nanomolar affinity was found (K d = 0.62 ± 0.33 nM) and applied to super-sensitive and red-emissive fluorescent staining of amyloid plaques in cortical brain tissue ex vivo. These fluorophores expand the dioxaborine-curcumin-based family of Aß-sensitive fluorophores with a promising new imaging agent.

15.
Nat Commun ; 13(1): 904, 2022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-35173167

RESUMEN

Targeted protein degradation allows targeting undruggable proteins for therapeutic applications as well as eliminating proteins of interest for research purposes. While several degraders that harness the proteasome or the lysosome have been developed, a technology that simultaneously degrades targets and accelerates cellular autophagic flux is still missing. In this study, we develop a general chemical tool and platform technology termed AUTOphagy-TArgeting Chimera (AUTOTAC), which employs bifunctional molecules composed of target-binding ligands linked to autophagy-targeting ligands. AUTOTACs bind the ZZ domain of the otherwise dormant autophagy receptor p62/Sequestosome-1/SQSTM1, which is activated into oligomeric bodies in complex with targets for their sequestration and degradation. We use AUTOTACs to degrade various oncoproteins and degradation-resistant aggregates in neurodegeneration at nanomolar DC50 values in vitro and in vivo. AUTOTAC provides a platform for selective proteolysis in basic research and drug development.


Asunto(s)
Autofagia/fisiología , Lisosomas/metabolismo , Proteínas Oncogénicas/metabolismo , Agregado de Proteínas/fisiología , Proteolisis , Línea Celular Tumoral , Células HeLa , Humanos , Unión Proteica/fisiología , Pliegue de Proteína , Proteostasis/fisiología , Transducción de Señal
16.
ACS Sens ; 6(6): 2281-2289, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34115933

RESUMEN

Tau aggregation is believed to have a strong association with the level of cognitive deficits in Alzheimer's disease (AD). Thus, optical brain imaging of tau aggregates has recently gained substantial attention as a promising tool for the early diagnosis of AD. However, selective imaging of tau aggregates is a major challenge due to sharing similar ß-sheet structures with homologous Aß fibrils. Herein, four quinoline-based fluorescent probes (Q-tau) were judiciously designed using the donor-acceptor architecture for selective imaging of tau aggregates. In particular, probe Q-tau 4 exhibited a strong intramolecular charge transfer and favorable photophysical profile, such as a large Stokes' shift and fluorescence emission wavelength of 630 nm in the presence of tau aggregates. The probe also displayed a "turn-on" fluorescence behavior toward tau fibrils with a 3.5-fold selectivity versus Aß fibrils. In addition, Q-tau 4 exhibited nanomolar binding affinity to tau aggregates (Kd = 16.6 nM), which was 1.4 times higher than that for Aß fibrils. The mechanism of "turn-on" fluorescence was proposed to be an environment-sensitive molecular rotor-like response. Moreover, ex vivo labeling of human AD brain sections demonstrated favorable colocalization of Q-tau 4 and the phosphorylated tau antibody, while comparable limited staining was observed with Aß fibrils. Molecular docking was conducted to obtain insights into the tau-binding mode of the probe. Collectively, Q-tau 4 has successfully been used as a tau-specific fluorescent imaging agent with lower background interference.


Asunto(s)
Enfermedad de Alzheimer , Quinolinas , Péptidos beta-Amiloides , Colorantes Fluorescentes , Humanos , Simulación del Acoplamiento Molecular , Proteínas tau
17.
J Neurosci ; 41(24): 5138-5156, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33972400

RESUMEN

Protein aggregation can induce explicit neurotoxic events that trigger a number of presently untreatable neurodegenerative disorders. Chaperones, on the other hand, play a neuroprotective role because of their ability to unfold and refold abnormal proteins. The progressive nature of neurotoxic events makes it important to discover endogenous factors that affect pathologic and molecular phenotypes of neurodegeneration in animal models. Here, we identified microtubule-associated protein tau, and chaperones Hsp70 (heat shock protein 70) and DNAJA1 (DJ2) as endogenous substrates of cereblon (CRBN), a substrate-recruiting subunit of cullin4-RING-E3-ligase. This recruitment results in ubiquitin-mediated degradation of tau, Hsp70, and DJ2. Knocking out CRBN enhances the chaperone activity of DJ2, resulting in decreased phosphorylation and aggregation of tau, improved association of tau with microtubules, and reduced accumulation of pathologic tau across brain. Functionally abundant DJ2 could prevent tau aggregation induced by various factors like okadaic acid and heparin. Depletion of CRBN also decreases the activity of tau-kinases including GSK3α/ß, ERK, and p38. Intriguingly, we found a high expression of CRBN and low levels of DJ2 in neuronal tissues of 5XFAD and APP knock-in male mouse models of Alzheimer's disease. This implies that CRBN-mediated DJ2/Hsp70 pathway may be compromised in neurodegeneration. Being one of the primary pathogenic events, elevated CRBN can be a contributing factor for tauopathies. Our data provide a functional link between CRBN and DJ2/Hsp70 chaperone machinery in abolishing the cytotoxicity of aggregation-prone tau and suggest that Crbn-/- mice serve as an animal model of resistance against tauopathies for further exploration of the molecular mechanisms of neurodegeneration.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Tauopatías/patología , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas tau/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Degeneración Nerviosa , Tauopatías/metabolismo
18.
J Enzyme Inhib Med Chem ; 36(1): 856-868, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33771089

RESUMEN

The present study describes evaluation of epigenetic regulation by a small molecule as the therapeutic potential for treatment of Huntington's disease (HD). We identified 5-allyloxy-2-(pyrrolidin-1-yl)quinoline (APQ) as a novel SETDB1/ESET inhibitor using a combined in silico and in vitro cell based screening system. APQ reduced SETDB1 activity and H3K9me3 levels in a HD cell line model. In particular, not only APQ reduced H3K9me3 levels in the striatum but it also improved motor function and neuropathological symptoms such as neuronal size and activity in HD transgenic (YAC128) mice with minimal toxicity. Using H3K9me3-ChIP and genome-wide sequencing, we also confirmed that APQ modulates H3K9me3-landscaped epigenomes in YAC128 mice. These data provide that APQ, a novel small molecule SETDB1 inhibitor, coordinates H3K9me-dependent heterochromatin remodelling and can be an epigenetic drug for treating HD, leading with hope in clinical trials of HD.


Asunto(s)
Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Heterocromatina/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Enfermedad de Huntington/tratamiento farmacológico , Neuronas/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Técnicas Biosensibles , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/química , Transferencia Resonante de Energía de Fluorescencia , Heterocromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , Estructura Molecular , Neuronas/metabolismo , Neuronas/patología
19.
ACS Chem Neurosci ; 11(19): 3144-3152, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-32915536

RESUMEN

Advanced understanding of Alzheimer's disease (AD) and several tauopathies over the past decades indicates the pathological importance of tau aggregation in these diseases. Herein, we demonstrated that adenosine triphosphate (ATP), a highly charged anionic molecule found abundantly in the cytosol of cells, catalyzes fibrillation of tau as well as human islet amyloid polypeptide, a representative of basic intrinsically disordered proteins. Our results showed that ATP attracts multiple lysine residues of the four-repeat domain of tau (K18) via supramolecular complexation, thereby forming dimers that are converted to nuclei and accelerate fibril elongation. However, ATP was not directly incorporated into the K18 fibrils, suggesting that ATP plays the role of a catalyst, rather than a reactant, during K18 fibrillation. We also characterized the correlation between ATP dyshomeostasis and tau aggregation in the cellular environment. Our multiple biophysical approaches, including native mass spectrometry (MS), small-angle X-ray scattering (SAXS), and molecular dynamics (MD) simulation, provided insights into the molecular-level influence of ATP on the structural changes and fibrillation of tau.


Asunto(s)
Amiloide , Proteínas tau , Adenosina Trifosfato , Humanos , Dispersión del Ángulo Pequeño , Difracción de Rayos X
20.
Prog Neurobiol ; : 101782, 2020 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-32105751

RESUMEN

Accumulation of abnormal tau aggregates in the brain is a pathological hallmark of multiple neurodegenerative disorders including Alzheimer's disease. Increasing evidence suggests that soluble tau aggregates play a key role in tau pathology as neurotoxic species causing neuronal cell death and act as prion-like seeds mediating tau propagation. Despite the pathological relevance, there is a paucity of methods to monitor tau oligomerization in the brain. As a tool to monitor tau self-assembly in the brain, we generated a novel tau transgenic mouse, named TauP301L-BiFC. By introducing bimolecular fluorescence complementation technique to human tau containing a P301L mutation, we were able to monitor and quantify tau self-assembly, represented by BiFC fluorescence in the brains of transgenic TauP301L-BiFC mice. TauP301L-BiFC mice showed soluble tau oligomerization from 3 months, showing significantly enriched BiFC fluorescence in the brain. Then, massive tau fragmentation occured at 6 months showing dramatically decreased TauP301L-BiFC fluorescence. The fragmented tau species served as a seed for insoluble tau aggregation. In a result, insoluble TauP301L-BiFC aggregates coaggregated with endogenous mouse tau accumulated in the brain, showing subsequently increased BiFC fluorescence from 9 months. Neuronal degeneration and cognitive deficits were observed from 12 months of age. TauP301L-BiFC mouse model demonstrated that methylene blue reduced the amount of soluble tau oligomers in the brain, resulting in the prevention of cognitive impairments. We assure that TauP301L-BiFC mice are a bona-fide animal tool to monitor pathological tau oligomerization in AD and other tauopathies.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA