Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Traffic ; 22(5): 153-170, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33527700

RESUMO

Alzheimer's disease (AD) is associated with age-related neurodegeneration, synaptic deformation and chronic inflammation mediated by microglia and infiltrated macrophages in the brain. Tau oligomers can be released from damaged neurons via various mechanisms such as exosomes, neurotransmitter, membrane leakage etc. Microglia sense the extracellular Tau through several cell-surface receptors and mediate chemotaxis and phagocytosis. The purinergic receptor P2Y12R recently gained interest in neurodegeneration for neuro-glial communication and microglial chemotaxis towards the site of plaque deposition. To understand the effect of extracellular Tau oligomers in microglial migration, the P2Y12R-mediated actin remodeling, reorientation of tubulin network and rate of migration were studied in the presence of ATP. The extracellular Tau species directly interacted with P2Y12R and also induced this purinoceptor expression in microglia. Microglial P2Y12R colocalized with remodeled membrane-associated actin network as a component of migration in response to Tau oligomers. As an inducer of P2Y12R, ATP facilitated the localization of P2Y12R in lamellipodia and filopodia during accelerated microglial migration. The direct interaction of extracellular Tau oligomers with microglial P2Y12R would facilitate the signal transduction in both way, directional chemotaxis and receptor-mediated phagocytosis. These unprecedented findings emphasize that microglia can modulate the membrane-associated actin structure and incorporate P2Y12R to perceive the axis and rate of chemotaxis in Tauopathy.


Assuntos
Actinas , Microglia , Quimiotaxia , Proteínas de Ligação ao GTP , Humanos , Microglia/metabolismo , Receptores Purinérgicos/metabolismo , Receptores Purinérgicos P2Y12
2.
Cell Physiol Biochem ; 57(2): 123-136, 2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-37074918

RESUMO

BACKGROUND/AIMS: Alzheimer's disease is a progressive neurological disorder characterized by the intracellular accumulation of Tau protein aggregates. In the present work, we studied the effect of Toluidine Blue and photo-excited Toluidine Blue on the aggregation of repeat Tau using in vitro assays. METHODS: The in vitro experiments were carried out on recombinant repeat Tau which was purified by cation exchange chromatography. The ThS fluorescence analysis was used to study the aggregation kinetics of Tau. CD spectroscopy and electron microscopy were used to study the secondary structure and morphology of Tau respectively. The actin cytoskeleton modulation was studied in Neuro2a cells with help of immunofluorescent microscopy. RESULTS: Results showed that Toluidine Blue efficiently inhibited the formation of higher-order aggregates, which was evidenced by Thioflavin S fluorescence assay, SDS-PAGE, and TEM. Immunofluorescence studies on the cytoskeleton of Neuro2a cells showed that Toluidine Blue and photo-excited Toluidine Blue treatment at a non-toxic concentration of 0.5 µM stimulated the formation of actin-rich lamellipodia and filopodia structures. Tubulin networks were also differentially modulated after the treatment of Toluidine Blue and photo-excited Toluidine Blue. End-binding protein 1 (EB1) levels were observed to increase after Toluidine Blue and photo-excited Toluidine Blue treatment indicating accelerated microtubule polymerization. CONCLUSION: The overall study suggested that Toluidine Blue inhibited the aggregation of soluble Tau and photo-excited Toluidine Blue disaggregated the pre-formed Tau filaments. In our study, TB and PE-TB were observed to be potent against Tau aggregation. We observed a distinctive modulation of actin, tubulin networks, and EB1 levels after TB and PE-TB treatment, which suggested that TB and PE-TB have potency against cytoskeleton deformities.


Assuntos
Doença de Alzheimer , Cloreto de Tolônio , Humanos , Cloreto de Tolônio/farmacologia , Tubulina (Proteína)/metabolismo , Actinas/metabolismo , Proteínas de Transporte , Citoesqueleto/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo
3.
J Neuroinflammation ; 20(1): 72, 2023 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-36915196

RESUMO

Microtubule-associated protein, Tau has been implicated in Alzheimer's disease for its detachment from microtubules and formation of insoluble intracellular aggregates within the neurons. Recent findings have suggested the expulsion of Tau seeds in the extracellular domain and their prion-like propagation between neurons. Transforming Growth Factor-ß1 (TGF-ß1) is a ubiquitously occurring cytokine reported to carry out immunomodulation and neuroprotection in the brain. TGF-ß-mediated regulation occurs at the level of neuronal survival and differentiation, glial activation (astrocyte and microglia), amyloid production-distribution-clearance and neurofibrillary tangle formation, all of which contributes to Alzheimer's pathophysiology. Its role in the reorganization of cytoskeletal architecture and remodelling of extracellular matrix to facilitate cellular migration has been well-documented. Microglia are the resident immune sentinels of the brain responsible for surveying the local microenvironment, migrating towards the beacon of pertinent damage and phagocytosing the cellular debris or patho-protein deposits at the site of insult. Channelizing microglia to target extracellular Tau could be a good strategy to combat the prion-like transmission and seeding problem in Alzheimer's disease. The current review focuses on reaffirming the role of TGF-ß1 signalling in Alzheimer's pathology and cytoskeletal reorganization and considers utilizing the approach of TGF-ß-triggered microglia-mediated targeting of extracellular patho-protein, Tau, as a possible potential strategy to combat Alzheimer's disease.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/patologia , Fator de Crescimento Transformador beta1/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta , Proteínas tau/metabolismo
4.
Cell Mol Neurobiol ; 41(6): 1175-1181, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32529542

RESUMO

Tau is a microtubule-associated protein with an intrinsically unstructured conformation. Tau is subjected to several pathological post-translational modifications (PTMs), leading to its loss of interaction with microtubules and accumulation as neurofibrillary tangles (NFTs) in neurons. Tau aggregates impede functions of endoplasmic reticulum and mitochondria leading to the generation of oxidative stress and in turn amplifying the Tau aggregation. Tau is channelled to chaperones for folding into their native form, which otherwise causes its degradation and clearance. Cellular response triggers the activation of ubiquitin-proteasome system or autophagy to facilitate Tau degradation, based on the PTMs or mutations associated with Tau. Further, autophagy can be selective where Hsc70 interacts with Tau in monomeric, oligomeric and aggregated form and drives its clearance by chaperone-mediated autophagy pathway (CMA). Lysosome-associated membrane proteins-2A (LAMP-2A) is the key player of CMA that recognises Hsc70-Tau complex and triggers the downstream cascade. Thus, it becomes challenging for mutant Tau to be cleared by CMA as it loses its affinity for Hsc70 and LAMP-2A. In such a scenario, Tau might be degraded by macroautophagy otherwise sequestered by aggresomes. Henceforth, the degradation of Tau and its blockage that is associated with various PTMs of Tau would explain the dynamics of Tau degradation or accumulation in AD. Further, unveiling the role of accessory proteins involved in these degradation pathways would help in understanding their loss of function and preventing Tau clearance.


Assuntos
Doença de Alzheimer/metabolismo , Autofagia/fisiologia , Agregados Proteicos/fisiologia , Transdução de Sinais/fisiologia , Proteínas tau/metabolismo , Doença de Alzheimer/patologia , Animais , Humanos , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Proteólise
5.
Cell Mol Neurobiol ; 41(4): 651-668, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32468440

RESUMO

Cholesterol, a principal constituent of the cell membrane, plays a crucial role in the brain by regulating the synaptic transmission, neuronal signaling, as well as neurodegenerative diseases. Defects in the cholesterol trafficking are associated with enhanced generation of hyperphosphorylated Tau and Amyloid-ß protein. Tau, a major microtubule-associated protein in the brain, is the key regulator of the mature neuron. Abnormally hyperphosphorylated Tau hampers the major functions related to microtubule assembly by promoting neurofibrillary tangles of paired helical filaments, twisted ribbons, and straight filaments. The observed pathological changes due to impaired cholesterol and Tau protein accumulation cause Alzheimer's disease. Thus, in order to regulate the pathogenesis of Alzheimer's disease, regulation of cholesterol metabolism, as well as Tau phosphorylation, is essential. The current review provides an overview of (1) cholesterol synthesis in the brain, neurons, astrocytes, and microglia; (2) the mechanism involved in modulating cholesterol concentration between the astrocytes and brain; (3) major mechanisms involved in the hyperphosphorylation of Tau and amyloid-ß protein; and (4) microglial involvement in its regulation. Thus, the answering key questions will provide an in-depth information on microglia involvement in managing the pathogenesis of cholesterol-modulated hyperphosphorylated Tau protein.


Assuntos
Doença de Alzheimer/metabolismo , Colesterol/metabolismo , Microglia/metabolismo , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Humanos , Tauopatias/metabolismo
6.
Cell Commun Signal ; 19(1): 28, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33627135

RESUMO

Alzheimer's disease is one of the neurodegenerative diseases, characterized by the accumulation of abnormal protein deposits, which disrupts signal transduction in neurons and other glia cells. The pathological protein in neurodegenerative diseases, Tau and amyloid-ß contribute to the disrupted microglial signaling pathways, actin cytoskeleton, and cellular receptor expression. The important secondary messenger lipids i.e., phosphatidylinositols are largely affected by protein deposits of amyloid-ß in Alzheimer's disease. Phosphatidylinositols are the product of different phosphatidylinositol kinases and the state of phosphorylation at D3, D4, and D5 positions of inositol ring. Phosphatidylinositol 3,4,5-triphosphate (PI 3, 4, 5-P3) involves in phagocytic cup formation, cell polarization, whereas Phosphatidylinositol 4,5-bisphosphate (PI 4, 5-P2)-mediates the process of phagosomes formation and further its fusion with early endosome.. The necessary activation of actin-binding proteins such as Rac, WAVE complex, and ARP2/3 complex for the actin polymerization in the process of phagocytosis, migration is regulated and maintained by PI 3, 4, 5-P3 and PI 4, 5-P2. The ratio and types of fatty acid intake can influence the intracellular secondary lipid messengers along with the cellular content of phaphatidylcholine and phosphatidylethanolamine. The Amyloid-ß deposits and extracellular Tau seeds disrupt phosphatidylinositides level and actin cytoskeletal network that hamper microglial-signaling pathways in AD. We hypothesize that being a lipid species intracellular levels of phosphatidylinositol would be regulated by dietary fatty acids. Further we are interested to understand phosphoinositide-based signaling cascades in phagocytosis and actin remodeling. Video Abstract.


Assuntos
Actinas/metabolismo , Doença de Alzheimer/metabolismo , Microglia/metabolismo , Fosfatidilinositóis/metabolismo , Animais , Humanos , Fagocitose , Transdução de Sinais
7.
Cell Commun Signal ; 19(1): 51, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33962636

RESUMO

Neurodegenerative diseases like Alzheimer's, Parkinson's and Huntington's disease involves abnormal aggregation and accumulation of toxic proteins aggregates. Post-translational modifications (PTMs) of the causative proteins play an important role in the etiology of disease as they could either slow down or accelerate the disease progression. Alzheimer disease is associated with the aggregation and accumulation of two major protein aggregates-intracellular neurofibrillary tangles made up of microtubule-associated protein Tau and extracellular Amyloid-ß plaques. Post-translational modifications are important for the regulation of Tau`s function but an imbalance in PTMs may lead to abnormal Tau function and aggregation. Tau methylation is one of the important PTM of Tau in its physiological state. However, the methylation signature on Tau lysine changes once it acquires pathological aggregated form. Tau methylation can compete with other PTMs such as acetylation and ubiquitination. The state of PTM at these sites determines the fate of Tau protein in terms of its function and stability. The global methylation in neurons, microglia and astrocytes are involved in multiple cellular functions involving their role in epigenetic regulation of gene expression via DNA methylation. Here, we have discussed the effect of methylation on Tau function in a site-specific manner and their cross-talk with other lysine modifications. We have also elaborated the role of methylation in epigenetic aspects and neurodegenerative conditions associated with the imbalance in methylation metabolism affecting global methylation state of cells. Video abstract.


Assuntos
Doença de Alzheimer/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Agregados Proteicos , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Epigênese Genética , Humanos , Metilação , Proteínas tau/química
8.
Cell Commun Signal ; 19(1): 16, 2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33579328

RESUMO

BACKGROUND: Amyloid aggregate deposition is the key feature of Alzheimer's disease. The proteinaceous aggregates found in the afflicted brain are the intra-neuronal neurofibrillary tangles formed by the microtubule-associated protein Tau and extracellular deposits, senile plaques, of amyloid beta (Aß) peptide proteolytically derived from the amyloid precursor protein. Accumulation of these aggregates has manifestations in the later stages of the disease, such as memory loss and cognitive inabilities originating from the neuronal dysfunction, neurodegeneration, and brain atrophy. Treatment of this disease at the late stages is difficult, and many clinical trials have failed. Hence, the goal is to find means capable of preventing the aggregation of these intrinsically disordered proteins by inhibiting the early stages of their pathological transformations. Polyphenols are known to be neuroprotective agents with the noticeable potential against many neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Prion diseases. METHODS: We analyzed the capability of Baicalein to inhibit aggregation of human Tau protein by a multifactorial analysis that included several biophysical and biochemical techniques. RESULTS: The potency of Baicalein, a polyphenol from the Scutellaria baicalensis Georgi, against in vitro Tau aggregation and PHF dissolution has been screened and validated. ThS fluorescence assay revealed the potent inhibitory activity of Baicalein, whereas ANS revealed its mechanism of Tau inhibition viz. by oligomer capture and dissociation. In addition, Baicalein dissolved the preformed mature fibrils of Tau thereby possessing a dual target action. Tau oligomers formed by Baicalein were non-toxic to neuronal cells, highlighting its role as a potent molecule to be screened against AD. CONCLUSION: In conclusion, Baicalein inhibits aggregation of hTau40 by enhancing the formation of SDS-stable oligomers and preventing fibril formation. Baicalein-induced oligomers do not affect the viability of the neuroblastoma cells. Therefore, Baicalein can be considered as a lead molecule against Tau pathology in AD. Video Abstract.


Assuntos
Flavanonas/farmacologia , Fármacos Neuroprotetores/farmacologia , Agregação Patológica de Proteínas/tratamento farmacológico , Proteínas tau/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Escherichia coli/genética , Heparina , Camundongos , Agregação Patológica de Proteínas/metabolismo , Conformação Proteica , Proteínas tau/química , Proteínas tau/genética
9.
Cell Commun Signal ; 19(1): 49, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33933071

RESUMO

BACKGROUND: Microtubule-associated protein Tau undergoes aggregation in Alzheimer`s disease (AD) and a group of other related diseases collectively known as Tauopathies. In AD, Tau forms aggregates, which are deposited intracellularly as neurofibrillary tangles. Histone deacetylase-6 (HDAC6) plays an important role in aggresome formation, where it recruits polyubiquitinated aggregates to the motor protein dynein. METHODS: Here, we have studied the effects of HDAC6 ZnF UBP on Tau phosphorylation, ApoE localization, GSK-3ß regulation and cytoskeletal organization in neuronal cells by immunocytochemical analysis. This analysis reveals that the cell exposure to the UBP-type zinc finger domain of HDAC6 (HDAC6 ZnF UBP) can modulate Tau phosphorylation and actin cytoskeleton organization. RESULTS: HDAC6 ZnF UBP treatment to cells did not affect their viability and resulted in enhanced neurite extension and formation of structures similar to podosomes, lamellipodia and podonuts suggesting the role of this domain in actin re-organization. Also, HDAC6 ZnF UBP treatment caused increase in nuclear localization of ApoE and tubulin localization in microtubule organizing centre (MTOC). Therefore, our studies suggest the regulatory role of this domain in different aspects of neurodegenerative diseases. Upon HDAC6 ZnF UBP treatment, inactive phosphorylated form of GSK-3ß increases without any change in total GSK-3ß level. CONCLUSIONS: HDAC6 ZnF UBP was found to be involved in cytoskeletal re-organization by modulating actin dynamics and tubulin localization. Overall, our study suggests that ZnF domain of HDAC6 performs various regulatory functions apart from its classical function in aggresome formation in protein misfolding diseases. Video abstract.


Assuntos
Actinas/metabolismo , Espaço Extracelular/enzimologia , Desacetilase 6 de Histona/química , Desacetilase 6 de Histona/metabolismo , Processamento de Proteína Pós-Traducional , Dedos de Zinco , Proteínas tau/metabolismo , Animais , Apolipoproteínas E/metabolismo , Apoptose , Linhagem Celular , Núcleo Celular/metabolismo , Regulação para Baixo , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Camundongos , Centro Organizador dos Microtúbulos/metabolismo , Modelos Biológicos , Neurônios/metabolismo , Fosforilação , Podossomos/metabolismo , Domínios Proteicos , Tubulina (Proteína)/metabolismo
10.
Biochemistry ; 59(48): 4546-4562, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33237772

RESUMO

Histone deacetylase 6 is a class II histone deacetylase primarily present in the cytoplasm and involved in the regulation of various cellular functions. It consists of two catalytic deacetylase domains and a unique zinc finger ubiquitin binding protein domain, which sets it apart from other HDACs. HDAC6 is known to regulate cellular activities by modifying the function of microtubules, HSP90, and cortactin through deacetylation. Apart from the catalytic activity of HDAC6, it interacts with other proteins through either the SE14 domain or the ZnF UBP domain to modulate their functions. Here, we have studied the role of the HDAC6 ZnF UBP domain as a modifier of Tau aggregation by its direct interaction with the polyproline region/repeat region of Tau. Interaction of HDAC6 ZnF UBP with Tau was found to reduce the propensity of Tau to self-aggregate and to disaggregate preformed aggregates in a concentration-dependent manner and also bring about the conformational changes in Tau protein. The interaction of HDAC6 ZnF UBP with Tau results in its degradation, suggesting either proteolytic activity of HDAC6 ZnF UBP or its role in enhancing autoproteolysis of Tau.


Assuntos
Desacetilase 6 de Histona/química , Desacetilase 6 de Histona/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Domínio Catalítico , Humanos , Técnicas In Vitro , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ressonância Magnética Nuclear Biomolecular , Agregados Proteicos , Ligação Proteica , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Estabilidade Proteica , Proteólise , Ubiquitina/química , Ubiquitina/metabolismo , Dedos de Zinco , Proteínas tau/ultraestrutura
11.
J Neuroinflammation ; 17(1): 93, 2020 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-32209097

RESUMO

Microglial polarization is an utmost important phenomenon in Alzheimer's disease that influences the brain environment. Polarization depends upon the types of responses that cells undergo, and it is characterized by receptors present on the cell surface and the secreted cytokines to the most. The expression of receptors on the surface is majorly influenced by internal and external factors such as dietary lipids. Types of fatty acids consumed through diet influence the brain environment and glial cell phenotype and types of receptors on microglia. Reports suggest that dietary habits influence microglial polarization and the switching of microglial phenotype is very important in neurodegenerative diseases. Omega-3 fatty acids have more influence on the brain, and they are found to regulate the inflammatory stage of microglia by fine-tuning the number of receptors expressed on microglia cells. In Alzheimer's disease, one of the pathological proteins involved is Tau protein, and microtubule-associated protein upon abnormal phosphorylation detaches from the microtubule and forms insoluble aggregates. Aggregated proteins have a tendency to propagate within the neurons and also become one of the causes of neuroinflammation. We hypothesize that tuning microglia towards anti-inflammatory phenotype would reduce the propagation of Tau in Alzheimer's disease.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Polaridade Celular/fisiologia , Gorduras na Dieta/metabolismo , Ácidos Graxos/metabolismo , Microglia/patologia , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Humanos , Microglia/metabolismo , Fagocitose/fisiologia , Fosforilação , Proteínas tau/metabolismo
12.
J Neuroinflammation ; 17(1): 10, 2020 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-31915009

RESUMO

BACKGROUND: Alzheimer's disease is associated with the accumulation of intracellular Tau tangles within neurons and extracellular amyloid-ß plaques in the brain parenchyma, which altogether results in synaptic loss and neurodegeneration. Extracellular concentrations of oligomers and aggregated proteins initiate microglial activation and convert their state of synaptic surveillance into a destructive inflammatory state. Although Tau oligomers have fleeting nature, they were shown to mediate neurotoxicity and microglial pro-inflammation. Due to the instability of oligomers, in vitro experiments become challenging, and hence, the stability of the full-length Tau oligomers is a major concern. METHODS: In this study, we have prepared and stabilized hTau40WT oligomers, which were purified by size-exclusion chromatography. The formation of the oligomers was confirmed by western blot, thioflavin-S, 8-anilinonaphthaalene-1-sulfonic acid fluorescence, and circular dichroism spectroscopy, which determine the intermolecular cross-ß sheet structure and hydrophobicity. The efficiency of N9 microglial cells to phagocytose hTau40WT oligomer and subsequent microglial activation was studied by immunofluorescence microscopy with apotome. The one-way ANOVA was performed for the statistical analysis of fluorometric assay and microscopic analysis. RESULTS: Full-length Tau oligomers were detected in heterogeneous globular structures ranging from 5 to 50 nm as observed by high-resolution transmission electron microscopy, which was further characterized by oligomer-specific A11 antibody. Immunocytochemistry studies for oligomer treatment were evidenced with A11+ Iba1high microglia, suggesting that the phagocytosis of extracellular Tau oligomers leads to microglial activation. Also, the microglia were observed with remodeled filopodia-like actin structures upon the exposure of oligomers and aggregated Tau. CONCLUSION: The peri-membrane polymerization of actin filament and co-localization of Iba1 relate to the microglial movements for phagocytosis. Here, these findings suggest that microglia modified actin cytoskeleton for phagocytosis and rapid clearance of Tau oligomers in Alzheimer's disease condition.


Assuntos
Actinas/metabolismo , Proteínas de Membrana/metabolismo , Microglia/metabolismo , Fagocitose/fisiologia , Animais , Linhagem Celular , Cromatografia em Gel/métodos , Humanos , Proteínas de Membrana/análise , Camundongos
13.
Cell Mol Life Sci ; 76(19): 3681-3694, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31093687

RESUMO

The prominent pathological consequences of Alzheimer's disease (AD) are the misfolding and mis-sorting of two cellular proteins, amyloid-ß and microtubule-associated protein Tau. The accumulation of toxic phosphorylated Tau inside the neurons induces the increased processing of amyloid-ß-associated signaling cascade and vice versa. Neuroinflammation-driven synaptic depletion and cognitive decline are substantiated by the cross talk of activated microglia and astroglia, leading to neuron degeneration. Microglia are the brain-resident immune effectors that prove their diverse functions in maintaining CNS homeostasis via collaboration with astrocytes and T lymphocytes. Age-related senescence and chronic inflammation activate microglia with increased pro-inflammatory markers, oxidative damage and phagocytosis. But the improper processing of misfolded protein via lysosomal pathway destines the spreading of 'seed' constituents to the nearby healthy neurons. Primed microglia process and present self-antigen such as amyloid-ß and modified Tau to the infiltrated T lymphocytes through MHC I/II molecules. After an effective conversation with CD4+ T cells, microglial phenotype can be altered from pro-active M1 to neuro-protective M2 type, which corresponds to the tissue remodeling and homeostasis. In this review, we are focusing on the change in functionality of microglia from innate to adaptive immune response in the context of neuroprotection, which may help in the search of novel immune therapy in AD.


Assuntos
Doença de Alzheimer/imunologia , Apresentação de Antígeno , Microglia/imunologia , Imunidade Adaptativa , Doença de Alzheimer/terapia , Encéfalo/imunologia , Humanos , Imunoterapia , Neuroimunomodulação , Dobramento de Proteína , Linfócitos T/imunologia
14.
Arch Biochem Biophys ; 676: 108153, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31622587

RESUMO

Ayurveda is the medicinal science, dealing with utilization of naturally available plant products for treatment. A wide variety of neuroprotective herbs have been reported in Ayurveda. Brahmi, Bacopa monnieri is a nootropic ayurvedic herb known to be effective in neurological disorders from ancient times. Numerous approaches including natural and synthetic compounds have been applied against Alzheimer's disease. Amyloid-ß and Tau are the hallmarks proteins of several neuronal dysfunctions resulting in Alzheimer's disease. Tau is a microtubule-associated protein known to be involved in progression of Alzheimer's disease. The generation of reaction oxygen species, increased neuroinflammation and neurotoxicity are the major physiological dysfunctions associated with Tau aggregates, which leads to dementia and behavioural deficits. Bacoside A, Bacoside B, Bacosaponins, Betulinic acid, etc; are the bioactive component of Brahmi belonging to various chemical families. Each chemical component known have its significant role in neuroprotection. The neuroprotective properties of Brahmi and its bioactive components including reduction of ROS, neuroinflammation, aggregation inhibition of Amyloid-ß and improvement of cognitive and learning behaviour. Here on basis of earlier studies we hypothesize the inhibitory role of Brahmi against Tau-mediated toxicity. The overall studies have concluded that Brahmi can be used as a lead formulation for treatment of Alzheimer's disease and other neurological disorders.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Bacopa/química , Ayurveda , Extratos Vegetais/farmacologia , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Humanos , Extratos Vegetais/uso terapêutico
15.
Arch Biochem Biophys ; 675: 108119, 2019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31568753

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder caused by protein misfolding, aggregation and accumulation in the brain. A large number of molecules are being screened against these pathogenic proteins but the focus for therapeutics is shifting towards the natural compounds as aggregation inhibitors, mainly due to their minimum adverse effects. Baicalein is a natural compound belonging to the class of flavonoids isolated from the Chinese herb Scutellaria baicalensis. Here we applied fluorescence, absorbance, microscopy, MALDI-TOF spectrophotometry and other biochemical techniques to investigate the interaction between Tau and Baicalein in vitro. We found the aggregation inhibitory properties of Baicalein for the repeat Tau. Overall, the potential of Baicalein in dissolving the preformed Tau oligomers as well as mature fibrils can be of utmost importance in therapeutics for Alzheimer's disease.


Assuntos
Doença de Alzheimer/metabolismo , Flavanonas/farmacologia , Proteínas tau/metabolismo , Linhagem Celular Tumoral , Humanos , Simulação de Acoplamento Molecular
16.
Proteomics ; 15(2-3): 245-59, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25315903

RESUMO

The receptor for advanced glycation end products (RAGE) is one of the most important proteins implicated in diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer. It is a pattern recognition receptor by virtue of its ability to interact with multiple ligands, RAGE activates several signal transduction pathways through involvement of various kinases that phosphorylate their respective substrates. Only few substrates have been known to be phosphorylated in response to activation by RAGE (e.g., nuclear factor kappa B); however, it is possible that these kinases can phosphorylate multiple substrates depending upon their expression and localization, leading to altered cellular responses in different cell types and conditions. One such example is, glycogen synthase kinase 3 beta which is known to phosphorylate glycogen synthase, acts downstream to RAGE, and hyperphosphorylates microtubule-associated protein tau causing neuronal damage. Thus, it is important to understand the role of various RAGE-activated kinases and their substrates. Therefore, we have reviewed here the details of RAGE-activated kinases in response to different ligands and their respective phosphoproteome. Furthermore, we discuss the analysis of the data mined for known substrates of these kinases from the PhosphoSitePlus (http://www.phosphosite.org) database, and the role of some of the important substrates involved in cancer, diabetes, cardiovascular diseases, and neurodegenerative diseases. In summary, this review provides information on RAGE-activated kinases and their phosphoproteome, which will be helpful in understanding the possible role of RAGE and its ligands in progression of diseases.


Assuntos
Proteínas Quinases/metabolismo , Proteômica/métodos , Receptores Imunológicos/metabolismo , Transdução de Sinais , Animais , Humanos , Fosforilação , Proteoma/metabolismo , Receptor para Produtos Finais de Glicação Avançada
17.
Hum Mol Genet ; 21(15): 3500-12, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22556362

RESUMO

Rare mutations in the gene encoding for tau (MAPT, microtubule-associated protein tau) cause frontotemporal dementia-spectrum (FTD-s) disorders, including FTD, progressive supranuclear palsy (PSP) and corticobasal syndrome, and a common extended haplotype spanning across the MAPT locus is associated with increased risk of PSP and Parkinson's disease. We identified a rare tau variant (p.A152T) in a patient with a clinical diagnosis of PSP and assessed its frequency in multiple independent series of patients with neurodegenerative conditions and controls, in a total of 15 369 subjects. Tau p.A152T significantly increases the risk for both FTD-s (n = 2139, OR = 3.0, CI: 1.6-5.6, P = 0.0005) and Alzheimer's disease (AD) (n = 3345, OR = 2.3, CI: 1.3-4.2, P = 0.004) compared with 9047 controls. Functionally, p.A152T (i) decreases the binding of tau to microtubules and therefore promotes microtubule assembly less efficiently; and (ii) reduces the tendency to form abnormal fibers. However, there is a pronounced increase in the formation of tau oligomers. Importantly, these findings suggest that other regions of the tau protein may be crucial in regulating normal function, as the p.A152 residue is distal to the domains considered responsible for microtubule interactions or aggregation. These data provide both the first genetic evidence and functional studies supporting the role of MAPT p.A152T as a rare risk factor for both FTD-s and AD and the concept that rare variants can increase the risk for relatively common, complex neurodegenerative diseases, but since no clear significance threshold for rare genetic variation has been established, some caution is warranted until the findings are further replicated.


Assuntos
Doença de Alzheimer/genética , Demência Frontotemporal/genética , Variação Genética , Proteínas tau/genética , Idoso , Doença de Alzheimer/epidemiologia , Demência Frontotemporal/epidemiologia , Predisposição Genética para Doença , Genótipo , Haplótipos , Humanos , Pessoa de Meia-Idade , Risco
18.
Adv Protein Chem Struct Biol ; 141: 467-493, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38960483

RESUMO

In Alzheimer's disease, the microtubule-associated protein, Tau misfolds to form aggregates and filaments in the intra- and extracellular region of neuronal cells. Microglial cells are the resident brain macrophage cells involved in constant surveillance and activated by the extracellular deposits. Purinergic receptors are involved in the chemotactic migration of microglial cells towards the site of inflammation. From our recent study, we have observed that the microglial P2Y12 receptor is involved in phagocytosis of full-length Tau species such as monomers, oligomers and aggregates by actin-driven chemotaxis. This study shows the interaction of repeat-domain of Tau (TauRD) with the microglial P2Y12 receptor and the corresponding residues for interaction have been analyzed by various in-silico approaches. In the cellular studies, TauRD was found to interact with microglial P2Y12R and induces its cellular expression confirmed by co-immunoprecipitation and western blot analysis. Furthermore, the P2Y12R-mediated TauRD internalization has demonstrated activation of microglia with an increase in the Iba1 level, and TauRD becomes accumulated at the peri-nuclear region for the degradation.


Assuntos
Tauopatias , Humanos , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/metabolismo , Microglia/metabolismo , Microglia/patologia , Receptores Purinérgicos P2Y12/metabolismo , Animais , Receptores Acoplados a Proteínas G/metabolismo
19.
Methods Mol Biol ; 2754: 105-116, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38512663

RESUMO

Tau aggregates are considered a pathological hallmark of Alzheimer's disease. The screening of molecules against Tau aggregation is a novel strategy for Alzheimer's disease. The photo-excited molecules have proven to be effective as a therapeutic agent in several diseases. In recent studies, the photo-excited dyes showed an inhibitory effect on Alzheimer's disease-related Tau protein aggregation and toxicity. The present chapter deals with the effect of rose bengal on the aggregation of Tau. The in vitro studies carried out with the help of electron microscopy, ThS fluorescence, and circular dichroism suggested that RB attenuated the Tau aggregation under in vitro conditions, whereas PE-RB disaggregated the mature Tau fibrils. Photo-excited rose bengal and the classical rose bengal induced a low degree of toxicity in cells. Thus, for the treatment of Alzheimer's disease, the rose bengal could be considered a potential molecule.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/metabolismo , Agregados Proteicos , Rosa Bengala/farmacologia , Rosa Bengala/uso terapêutico , Corantes , Proteínas tau/metabolismo , Microscopia Eletrônica , Agregação Patológica de Proteínas/metabolismo
20.
Adv Protein Chem Struct Biol ; 141: 447-466, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38960482

RESUMO

Alzheimer's disease is progressive neurodegenerative disease characterize by the presence of extracellular accumulation of amyloid-ß plaques and intracellular deposits of neurofibrillary tangles of Tau. Apart from axonal depositions pathological aggregated Tau protein is known to secrete into extracellular spaces and propagate through seeding mechanism. Microglia, the immune cells of the brain display modest ability to internalize the extracellular Tau and degrade it through endolysosomal pathway. However, the excessive burden of pathoproteins weakens the phagocytic ability of microglia. Extracellular supplementation of omega-3 fatty acids (n-3) may regulate the phagocytosis of microglia as they mediate the anti-inflammatory polarization of microglia through membrane lipid compositions changes. The internalization of extracellular Tau in the microglia is regulated by cortical membrane-associated actin remodeling driven by interplay of actin-binding proteins. On the other hand, Tau display capability bind and interact with various actin-binding protein owing to the presence of proline-rich domain in the structure and regulate their activation. In this study, we hypothesize that internalization of Tau in the presence of omega-3 fatty acids would propagate the Tau-mediated activation of actin-binding proteins as well as extracellular matrix and in turn modulate cortical actin remodeling for phagocytosis.


Assuntos
Proteínas da Matriz Extracelular , Proteínas tau , Proteínas tau/metabolismo , Humanos , Proteínas da Matriz Extracelular/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Fagocitose , Animais , Ácidos Graxos Ômega-3/metabolismo , Microglia/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa