Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 280
Filtrar
1.
J Struct Biol ; 216(2): 108092, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38615725

RESUMO

Cerebral amyloid angiopathy (CAA) is associated with the accumulation of fibrillar Aß peptides upon and within the cerebral vasculature, which leads to loss of vascular integrity and contributes to disease progression in Alzheimer's disease (AD). We investigate the structure of human-derived Aß40 fibrils obtained from patients diagnosed with sporadic or familial Dutch-type (E22Q) CAA. Using cryo-EM, two primary structures are identified containing elements that have not been observed in in vitro Aß40 fibril structures. One population has an ordered N-terminal fold comprised of two ß-strands stabilized by electrostatic interactions involving D1, E22, D23 and K28. This charged cluster is disrupted in the second population, which exhibits a disordered N-terminus and is favored in fibrils derived from the familial Dutch-type CAA patient. These results illustrate differences between human-derived CAA and AD fibrils, and how familial CAA mutations can guide fibril formation.


Assuntos
Peptídeos beta-Amiloides , Angiopatia Amiloide Cerebral , Eletricidade Estática , Humanos , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/química , Angiopatia Amiloide Cerebral/patologia , Angiopatia Amiloide Cerebral/genética , Angiopatia Amiloide Cerebral/metabolismo , Microscopia Crioeletrônica/métodos , Amiloide/metabolismo , Amiloide/química , Amiloide/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Mutação , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo
2.
J Biol Chem ; 299(4): 103027, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36805335

RESUMO

Imbalances in the amounts of amyloid-ß peptides (Aß) generated by the membrane proteases ß- and γ-secretase are considered as a trigger of Alzheimer's disease (AD). Cell-free studies of γ-secretase have shown that increasing membrane thickness modulates Aß generation but it has remained unclear if these effects are translatable to cells. Here we show that the very long-chain fatty acid erucic acid (EA) triggers acyl chain remodeling in AD cell models, resulting in substantial lipidome alterations which included increased esterification of EA in membrane lipids. Membrane remodeling enhanced γ-secretase processivity, resulting in the increased production of the potentially beneficial Aß37 and/or Aß38 species in multiple cell lines. Unexpectedly, we found that the membrane remodeling stimulated total Aß secretion by cells expressing WT γ-secretase but lowered it for cells expressing an aggressive familial AD mutant γ-secretase. We conclude that EA-mediated modulation of membrane composition is accompanied by complex lipid homeostatic changes that can impact amyloidogenic processing in different ways and elicit distinct γ-secretase responses, providing critical implications for lipid-based AD treatment strategies.


Assuntos
Doença de Alzheimer , Secretases da Proteína Precursora do Amiloide , Humanos , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Lipídeos de Membrana/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Linhagem Celular , Precursor de Proteína beta-Amiloide/metabolismo , Presenilina-1/metabolismo
3.
Chemistry ; 30(48): e202400277, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-38888453

RESUMO

Amyloid plaques are a major pathological hallmark involved in Alzheimer's disease and consist of deposits of the amyloid-ß peptide (Aß). The aggregation process of Aß is highly complex, which leads to polymorphous aggregates with different structures. In addition to aberrant aggregation, Aß oligomers can undergo liquid-liquid phase separation (LLPS) and form dynamic condensates. It has been hypothesized that these amyloid liquid droplets affect and modulate amyloid fibril formation. In this review, we briefly introduce the relationship between stress granules and amyloid protein aggregation that is associated with neurodegenerative diseases. Then we highlight the regulatory role of LLPS in Aß aggregation and discuss the potential relationship between Aß phase transition and aggregation. Furthermore, we summarize the current structures of Aß oligomers and amyloid fibrils, which have been determined using nuclear magnetic resonance (NMR) and cryo-electron microscopy (cryo-EM). The structural variations of Aß aggregates provide an explanation for the different levels of toxicity, shed light on the aggregation mechanism and may pave the way towards structure-based drug design for both clinical diagnosis and treatment.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Humanos , Microscopia Crioeletrônica , Agregados Proteicos , Amiloide/química , Amiloide/metabolismo , Agregação Patológica de Proteínas/metabolismo
4.
FASEB J ; 37(5): e22890, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37002885

RESUMO

Amyloid-ß (Aß) peptide is accumulated in the mitochondria and has been shown to play a central role in the development of Alzheimer's disease (AD). It has been shown that exposure of neurons to aggregated Aß can result in damaged mitochondria and dysregulated mitophagy, indicating that changes in the Aß content of mitochondria may affect the levels of mitophagy and interfere with the progression of AD. However, the direct influence of mitochondrial Aß on mitophagy has not been elucidated. In the present study, the effect of the mitochondria-specific Aß was assessed following a direct change of Aß content in the mitochondria. We directly change mitochondrial Aß by transfecting cells with mitochondria-associated plasmids, including the mitochondrial outer membrane protein translocase 22 (TOMM22) and 40 (TOMM40) or presequence protease (PreP) overexpression plasmids. The changes in the levels of mitophagy were assessed by TEM, Western blot, mito-Keima construct, organelle tracker, and probe JC-1 assay. We demonstrated that increased mitochondrial Aß content enhance mitophagy levels; overexpression of PreP could reverse the mitochondrial Aß-induced mitophagy levels in vivo and in vitro by reversing the levels of reactive oxygen species (ROS) and the mitochondrial membrane potential. The data provide novel insight into the role of mitochondria-specific Aß in the progression of AD pathophysiology.


Assuntos
Doença de Alzheimer , Mitofagia , Humanos , Peptídeos beta-Amiloides/metabolismo , Mitocôndrias/metabolismo , Doença de Alzheimer/metabolismo , Peptídeo Hidrolases/metabolismo
5.
Bioessays ; 44(11): e2200086, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36104212

RESUMO

Amyloid fibril formation plays a central role in the pathogenesis of a number of neurodegenerative diseases, including Alzheimer and Parkinson diseases. Transient prefibrillar oligomers forming during the aggregation process, exhibiting a small size and a large hydrophobic surface, can aberrantly interact with a number of molecular targets on neurons, including the lipid bilayer of plasma membranes, resulting in a fatal outcome for the cells. By contrast, the mature fibrils, despite presenting generally a high hydrophobic surface, are endowed with a low diffusion rate and poorly penetrate the interior of the lipid bilayer. However, increasing evidence shows that both intracellular α-synuclein fibrils, as well and as extracellular amyloid-ß and ß2-microglobulin fibrils, can release oligomers over time that quickly diffuse to reach the membrane of the neighboring cells. The persistent leakage of harmful oligomers from fibrils triggers an ongoing cascade of events resulting in a sustained injury to neurons and glia and also provides aggregates with the ability to cross biological membranes and diffuse between cells or cellular compartments.


Assuntos
Amiloide , Doença de Parkinson , Humanos , Amiloide/química , Amiloide/metabolismo , alfa-Sinucleína/metabolismo , Bicamadas Lipídicas , Peptídeos beta-Amiloides/metabolismo , Doença de Parkinson/metabolismo
6.
Biochem Soc Trans ; 51(1): 147-159, 2023 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-36629697

RESUMO

Misfolding, aggregation and accumulation of Amyloid-ß peptides (Aß) in neuronal tissue and extracellular matrix are hallmark features of Alzheimer's disease (AD) pathology. Soluble Aß oligomers are involved in neuronal toxicity by interacting with the lipid membrane, compromising its integrity, and affecting the function of receptors. These facts indicate that the interaction between Aß oligomers and cell membranes may be one of the central molecular level factors responsible for the onset of neurodegeneration. The present review provides a structural understanding of Aß neurotoxicity via membrane interactions and contributes to understanding early events in Alzheimer's disease.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Humanos , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/metabolismo , Fosfolipídeos/metabolismo , Membrana Celular/metabolismo , Amiloide/metabolismo
7.
Int J Neurosci ; 133(8): 888-895, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34818135

RESUMO

PURPOSE: Although amyloid-ß (Aß) is one of the neuropathological hallmarks of Alzheimer's Disease (AD), the mechanisms of Aß neurotoxicity remain to be clarified. This study was aimed to evaluate the effect of Aß on postsynaptic density-95 (PSD-95) tyrosine phosphorylation. Elucidating the regulatory mechanisms underlying it may be a promising therapy in AD. METHODS: Aß25-35 oligomers (20 µg/rat) were administered intracerebroventricularly in adult male Sprague-Dawley rats. PSD-95 tyrosine phosphorylation was assessed using immunoprecipitation followed by immunoblot analysis. Immunoblot was applied for measuring the protein levels of PSD-95 and ß-actin. RESULTS: Following 3, 7, 14, 21 days after oligomeric Aß25-35 treatment, the tyrosine phosphorylation of PSD-95 increased significantly, and peaked at 3 days after oligomeric Aß25-35 treatment in hippocampal CA1 subfield. Src family protein tyrosine kinases (SrcPTKs) specific inhibitor PP2 attenuated the tyrosine phosphorylation of PSD-95 induced by Aß25-35. Amantadine [N-methyl-D-aspartate (NMDA) receptor noncompetitive antagonist], NVP-AAM077 (GluN2A-containing NMDA receptor selective inhibitor) and Ro25-6981 (GluN2B-containing NMDA receptor selective inhibitor) also suppressed the Aß25-35-induced PSD-95 tyrosine phosphorylation. CONCLUSION: These results suggest that Aß oligomers induce the tyrosine phosphorylation of PSD-95 by SrcPTKs, which is mediated by the activation of GluN2A- and GluN2B-containing NMDA receptors.


Assuntos
Doença de Alzheimer , Receptores de N-Metil-D-Aspartato , Animais , Masculino , Ratos , Doença de Alzheimer/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Hipocampo/metabolismo , Fosforilação , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Fatores de Transcrição/metabolismo , Tirosina/metabolismo
8.
Drug Dev Res ; 84(8): 1578-1594, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37675624

RESUMO

Alzheimer's disease (AD) is a chronic and irreversible neurodegenerative disease associated with aging. It is characterized by the progressive loss of memory and other cognitive functions. Although the exact etiology of AD is not well explored, several factors, such as the deposition of amyloid-ß (Aß) plaques, hyperphosphorylation of tau protein, presence of low levels of acetylcholine, and generation of oxidative stress, are key mediators in the progression of AD. Currently, the clinical treatment options for AD are limited and are based on cholinesterase (ChE) inhibitors (e.g., donepezil, rivastigmine, and galantamine), N-methyl- d-aspartic acid receptor antagonists (e.g., memantine), and the recently approved Aß modulator (e.g., aducanumab). Tryptamine (2-(1H-indol-3-yl)ethan-1-amine) is a small molecule that contains an indole nucleus and an ethylamine side chain. It is also the active metabolite of tryptophan. It possesses a wide range of biological activities related to neurodegenerative disorders, such as ChE inhibition, Aß aggregation inhibition, antioxidant effects, monoamine-oxidase inhibition, and neuroprotection. Several tryptamine-based hybrid analogs are currently being investigated as multifunctional agents for the development of novel hybrids for AD treatment. Thus, this review article aims to provide in-depth insights into the research progress and strategies for designing multifunctional agents used in Alzheimer's therapy.


Assuntos
Doença de Alzheimer , Doenças Neurodegenerativas , Humanos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico , Inibidores da Colinesterase/farmacologia , Donepezila , Peptídeos beta-Amiloides , Triptaminas/farmacologia , Triptaminas/uso terapêutico
9.
Int J Mol Sci ; 24(21)2023 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-37958934

RESUMO

Alzheimer's disease (AD) is the most common neurodegenerative disorder and the main cause of dementia which is characterized by a progressive cognitive decline that severely interferes with daily activities of personal life. At a pathological level, it is characterized by the accumulation of abnormal protein structures in the brain-ß-amyloid (Aß) plaques and Tau tangles-which interfere with communication between neurons and lead to their dysfunction and death. In recent years, research on AD has highlighted the critical involvement of mitochondria-the primary energy suppliers for our cells-in the onset and progression of the disease, since mitochondrial bioenergetic deficits precede the beginning of the disease and mitochondria are very sensitive to Aß toxicity. On the other hand, if it is true that the accumulation of Aß in the mitochondria leads to mitochondrial malfunctions, it is otherwise proven that mitochondrial dysfunction, through the generation of reactive oxygen species, causes an increase in Aß production, by initiating a vicious cycle: there is therefore a bidirectional relationship between Aß aggregation and mitochondrial dysfunction. Here, we focus on the latest news-but also on neglected evidence from the past-concerning the interplay between dysfunctional mitochondrial complex I, oxidative stress, and Aß, in order to understand how their interplay is implicated in the pathogenesis of the disease.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Mitocôndrias/metabolismo
10.
Int J Mol Sci ; 24(12)2023 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-37372938

RESUMO

Alzheimer's disease (AD) is an age-related, multifaceted neurological disorder associated with accumulation of aggregated proteins (amyloid Aß and hyperphosphorylated tau), loss of synapses and neurons, and alterations in microglia. AD was recognized by the World Health Organization as a global public health priority. The pursuit of a better understanding of AD forced researchers to pay attention to well-defined single-celled yeasts. Yeasts, despite obvious limitations in application to neuroscience, show high preservation of basic biological processes with all eukaryotic organisms and offer great advantages over other disease models due to the simplicity, high growth rates on low-cost substrates, relatively simple genetic manipulations, the large knowledge base and data collections, and availability of an unprecedented amount of genomic and proteomic toolboxes and high-throughput screening techniques, inaccessible to higher organisms. Research reviewed above clearly indicates that yeast models, together with other, more simple eukaryotic models including animal models, C. elegans and Drosophila, significantly contributed to understanding Aß and tau biology. These models allowed high throughput screening of factors and drugs that interfere with Aß oligomerization, aggregation and toxicity, and tau hyperphosphorylation. In the future, yeast models will remain relevant, with a focus on creating novel high throughput systems to facilitate the identification of the earliest AD biomarkers among different cellular networks in order to achieve the main goal-to develop new promising therapeutic strategies to treat or prevent the disease.


Assuntos
Doença de Alzheimer , Animais , Doença de Alzheimer/metabolismo , Saccharomyces cerevisiae/metabolismo , Peptídeos beta-Amiloides/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Caenorhabditis elegans/metabolismo , Proteômica , Modelos Animais de Doenças
11.
Int J Mol Sci ; 24(11)2023 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-37298510

RESUMO

Alzheimer's disease (AD) is one of the most common neurodegenerative diseases. In AD patients, amyloid-ß (Aß) peptide-mediated degeneration of the cholinergic system utilizing acetylcholine (ACh) for memory acquisition is observed. Since AD therapy using acetylcholinesterase (AChE) inhibitors are only palliative for memory deficits without reversing disease progress, there is a need for effective therapies, and cell-based therapeutic approaches should fulfil this requirement. We established F3.ChAT human neural stem cells (NSCs) encoding the choline acetyltransferase (ChAT) gene, an ACh-synthesizing enzyme, HMO6.NEP human microglial cells encoding the neprilysin (NEP) gene, an Aß-degrading enzyme, and HMO6.SRA cells encoding the scavenger receptor A (SRA) gene, an Aß-uptaking receptor. For the efficacy evaluation of the cells, first, we established an appropriate animal model based on Aß accumulation and cognitive dysfunction. Among various AD models, intracerebroventricular (ICV) injection of ethylcholine mustard azirinium ion (AF64A) induced the most severe Aß accumulation and memory dysfunction. Established NSCs and HMO6 cells were transplanted ICV to mice showing memory loss induced by AF64A challenge, and brain Aß accumulation, ACh concentration and cognitive function were analyzed. All the transplanted F3.ChAT, HMO6.NEP and HMO6.SRA cells were found to survive up to 4 weeks in the mouse brain and expressed their functional genes. Combinational treatment with the NSCs (F3.ChAT) and microglial cells encoding each functional gene (HMO6.NEP or HMO6.SRA) synergistically restored the learning and memory function of AF64A-challenged mice by eliminating Aß deposits and recovering ACh level. The cells also attenuated inflammatory astrocytic (glial fibrillary acidic protein) response by reducing Aß accumulation. Taken together, it is expected that NSCs and microglial cells over-expressing ChAT, NEP or SRA genes could be strategies for replacement cell therapy of AD.


Assuntos
Doença de Alzheimer , Células-Tronco Neurais , Humanos , Camundongos , Animais , Doença de Alzheimer/genética , Doença de Alzheimer/terapia , Doença de Alzheimer/metabolismo , Microglia/metabolismo , Acetilcolinesterase/metabolismo , Células-Tronco Neurais/metabolismo , Peptídeos beta-Amiloides/metabolismo , Transtornos da Memória/metabolismo , Neprilisina/metabolismo , Acetilcolina/metabolismo , Modelos Animais de Doenças
12.
Neurochem Res ; 47(3): 748-761, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34783973

RESUMO

Hydroxysafflor yellow A (HSYA) is an extract from Carthamus tinctorius L. dry flowers (Compositae). HSYA has been shown to have neuroprotective effects on several Alzheimer's disease (AD) models. However, the exact mechanisms by which HSYA regulates neuroinflammation have still not been clarified. In this study, we investigated the mechanism by which HSYA regulates microglial activation and neuroinflammation via TREM2, and further clarified its underlying molecular mechanism. We silenced TREM2 in BV-2 cells and evaluated the expression of inflammatory markers (TNF-α, IL-1ß, IL-4, IL-6, IL-10, and IL-13). The results showed that HSYA could up-regulate cell viability and improve the morphology of BV-2 cells injured by Aß1-42. The results showed that Aß1-42 could induce microglia to upregulate the expression of M1 markers (iNOS, IL-1ß, IL-6) and downregulate M2 marker (Arg-1, IL-4, IL-10, IL-13) expression. HSYA reversed the effects of Aß1-42 via TREM2, switching microglia from an M1 proinflammatory phenotype to an M2 anti-inflammatory phenotype. HSYA inhibited the Aß1-42-induced activation of the TLR4/NF-κB transduction pathway by upregulating TREM2 and regulated the transcription of inflammatory cytokines via the downstream transcription factors NF-κB p65 and IκB-α. In conclusion, HSYA regulated the microglial inflammatory phenotype by regulating microglial (M1/M2) polarization in Aß1-42-induced BV-2 cells which may be mediated by the TREM2/TLR4/NF-κB pathway.


Assuntos
Microglia , NF-kappa B , Peptídeos beta-Amiloides , Animais , Chalcona/análogos & derivados , Glicoproteínas de Membrana/metabolismo , Camundongos , Microglia/metabolismo , NF-kappa B/metabolismo , Doenças Neuroinflamatórias , Fragmentos de Peptídeos , Fenótipo , Quinonas , Receptores Imunológicos/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
13.
Int J Mol Sci ; 23(18)2022 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-36142143

RESUMO

The blood-brain barrier (BBB) is a selective barrier and a functional gatekeeper for the central nervous system (CNS), essential for maintaining brain homeostasis. The BBB is composed of specialized brain endothelial cells (BECs) lining the brain capillaries. The tight junctions formed by BECs regulate paracellular transport, whereas transcellular transport is regulated by specialized transporters, pumps and receptors. Cytokine-induced neuroinflammation, such as the tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß), appear to play a role in BBB dysfunction and contribute to the progression of Alzheimer's disease (AD) by contributing to amyloid-ß (Aß) peptide accumulation. Here, we investigated whether TNF-α and IL-1ß modulate the permeability of the BBB and alter Aß peptide transport across BECs. We used a human BBB in vitro model based on the use of brain-like endothelial cells (BLECs) obtained from endothelial cells derived from CD34+ stem cells cocultivated with brain pericytes. We demonstrated that TNF-α and IL-1ß differentially induced changes in BLECs' permeability by inducing alterations in the organization of junctional complexes as well as in transcelluar trafficking. Further, TNF-α and IL-1ß act directly on BLECs by decreasing LRP1 and BCRP protein expression as well as the specific efflux of Aß peptide. These results provide mechanisms by which CNS inflammation might modulate BBB permeability and promote Aß peptide accumulation. A future therapeutic intervention targeting vascular inflammation at the BBB may have the therapeutic potential to slow down the progression of AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Interleucina-1beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Humanos , Inflamação/metabolismo , Proteínas de Neoplasias/metabolismo , Permeabilidade
14.
Int J Mol Sci ; 23(11)2022 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-35682848

RESUMO

The deposition of amyloid-ß peptide (Aß) in the brain is a critical event in the progression of Alzheimer's disease (AD). This Aß deposition could be prevented by directed enhancement of Aß binding to its natural depot, human serum albumin (HSA). Previously, we revealed that specific endogenous ligands of HSA improve its affinity to monomeric Aß. We show here that an exogenous HSA ligand, ibuprofen (IBU), exerts the analogous effect. Plasmon resonance spectroscopy data evidence that a therapeutic IBU level increases HSA affinity to monomeric Aß40/Aß42 by a factor of 3-5. Using thioflavin T fluorescence assay and transmission electron microcopy, we show that IBU favors the suppression of Aß40 fibrillation by HSA. Molecular docking data indicate partial overlap between the IBU/Aß40-binding sites of HSA. The revealed enhancement of the HSA-Aß interaction by IBU and the strengthened inhibition of Aß fibrillation by HSA in the presence of IBU could contribute to the neuroprotective effects of the latter, previously observed in mouse and human studies of AD.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Ibuprofeno/farmacologia , Ibuprofeno/uso terapêutico , Ligantes , Camundongos , Simulação de Acoplamento Molecular , Fragmentos de Peptídeos/metabolismo , Albumina Sérica/metabolismo , Albumina Sérica Humana
15.
J Membr Biol ; 254(5-6): 463-473, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34327545

RESUMO

Amyloid-ß peptide (Aß) has been shown to cause synaptic dysfunction and can render neurons vulnerable to excitotoxicity and oxidative stress. Na,K-ATPase plays an important role to maintain cell ionic equilibrium and it can be modulated by N-methyl-D-aspartate (NMDA)-nitric oxide (NO)-cyclic GMP pathway. Disruption of NO synthase (NOS) activity and reactive oxygen species (ROS) production could lead to changes in Na,K-ATPase isoforms' activities that may be detrimental to the cells. Our aim was to evaluate the signaling pathways of Aß in relation to NMDA-NOS-cyclic GMP versus oxidative stress on α1-/α2,3-Na,K-ATPase activities in rat hippocampal slices. Aß1-40 induced a concentration-dependent increase of NOS activity and increased cyclic guanosine monophosphate (cGMP), TBARS (thiobarbituric acid reactive substances), and 3-Nitrotyrosine (3-NT)-modified protein levels in rat hippocampal slices. The increase in NOS activity and cyclic GMP levels induced by Aß1-40 was completely blocked by MK-801 (inhibitor of NMDA receptor) and L-NAME (inhibitor of NOS) pre-treatment but changes in TBARS levels were only partially blocked by both compounds. The Aß treatment also decreased Na,K-ATPase activity which was reverted by N-nitro-L-arginine methyl ester hydrochloride (L-NAME) but not by MK-801 pre-treatment. The decrease in enzyme activity induced by Aß was isoform-specific since only α1-Na,K-ATPase was affected. These findings suggest that the activation of NMDA-NOS signaling cascade linked to α2,3-Na,K-ATPase activity may mediate an adaptive, neuroprotective response to Aß in rat hippocampus.


Assuntos
Hipocampo , Estresse Oxidativo , Animais , GMP Cíclico , Maleato de Dizocilpina , N-Metilaspartato , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico , Peptídeos , Ratos , ATPase Trocadora de Sódio-Potássio , Substâncias Reativas com Ácido Tiobarbitúrico
16.
Prostaglandins Other Lipid Mediat ; 156: 106582, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34273491

RESUMO

Millions of people are affected by neurodegenerative diseases worldwide. They occur due to the loss of brain functions or peripheral nervous system dysfunction. If untreated, prolonged condition ultimately leads to death. Mostly they are associated with stress, altered cholesterol metabolism, inflammation and organelle dysfunction. Endogenous cholesterol and phospholipids in brain undergo auto-oxidation by enzymatic as well as non-enzymatic modes leading to the formation of by-products such as 4-hydroxynonenal and oxysterols. Among various oxysterols, 7-ketocholesterol (7KCh) is one of the major toxic components involved in altering neuronal lipid metabolism, contributing to inflammation and nerve cell damage. More evidently 7KCh is proven to induce oxidative stress and affects membrane permeability. Loss in mitochondrial membrane potential affects metabolism of cell organelles such as lysosomes and peroxisomes which are involved in lipid and protein homeostasis. This in turn could affect amyloidogenesis, tau protein phosphorylation and accumulation in pathological conditions of neurodegenerative diseases. Lipid alterations and the consequent pathogenic protein accumulation, results in the damage of cell organelles and microglial cells. This could be a reason behind disease progression and predominantly reported characteristics of neurodegenerative disorders such as Alzheimer's disease. This review focuses on the role of 7KCh mediated neurodegenerative Alzheimer's disease with emphasis on alterations in the lipid raft microdomain. In addition, current trends in the significant therapies related to 7KCh inhibition are highlighted.


Assuntos
Doença de Alzheimer
17.
Mikrochim Acta ; 188(2): 56, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33502585

RESUMO

A sensitive and rapid colorimetric biosensor has been developed for determination of amyloid-ß peptide (Aß) and study of amyloidogenesis based on the high peroxidase-like activity of porous bimetallic ZnO-Co3O4 nanocages (NCs). Due to the high binding ability of Aß monomer to ZnO-Co3O4 NCs, the catalytic activity of ZnO-Co3O4 NCs can be significantly suppressed by Aß monomer. This finding forms the basis for a colorimetric assay for Aß monomer detection. The detection limit for Aß monomer is 3.5 nM with a linear range of 5 to 150 nM (R2 = 0.997). The system was successfully applied to the determination of Aß monomer in rat cerebrospinal fluid. Critically, the different inhibition effects of monomeric and aggregated Aß species on the catalytic activity of ZnO-Co3O4 NCs enabled the sensor to be used for tracking the dynamic progress of Aß aggregation and screening Aß inhibitors. Compared with the commonly used thioflavin T fluorescence assay, this method provided higher sensitivity to the formation of Aß oligomer at the very early assembly stage. Our assay shows potential application in early diagnosis and therapy of Alzheimer's disease (AD).


Assuntos
Peptídeos beta-Amiloides/líquido cefalorraquidiano , Colorimetria/métodos , Estruturas Metalorgânicas/química , Fragmentos de Peptídeos/líquido cefalorraquidiano , Animais , Benzidinas/química , Catálise , Compostos Cromogênicos/química , Cobalto/química , Limite de Detecção , Óxidos/química , Porosidade , Ratos , Óxido de Zinco/química
18.
Sensors (Basel) ; 21(6)2021 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-33809416

RESUMO

Alzheimer's disease (AD), considered a common type of dementia, is mainly characterized by a progressive loss of memory and cognitive functions. Although its cause is multifactorial, it has been associated with the accumulation of toxic aggregates of the amyloid-ß peptide (Aß) and neurofibrillary tangles (NFTs) of tau protein. At present, the development of highly sensitive, high cost-effective, and non-invasive diagnostic tools for AD remains a challenge. In the last decades, nanomaterials have emerged as an interesting and useful tool in nanomedicine for diagnostics and therapy. In particular, plasmonic nanoparticles are well-known to display unique optical properties derived from their localized surface plasmon resonance (LSPR), allowing their use as transducers in various sensing configurations and enhancing detection sensitivity. Herein, this review focuses on current advances in in vitro sensing techniques such as Surface-enhanced Raman scattering (SERS), Surface-enhanced fluorescence (SEF), colorimetric, and LSPR using plasmonic nanoparticles for improving the sensitivity in the detection of main biomarkers related to AD in body fluids. Additionally, we refer to the use of plasmonic nanoparticles for in vivo imaging studies in AD.


Assuntos
Doença de Alzheimer , Nanopartículas Metálicas , Doença de Alzheimer/diagnóstico , Peptídeos beta-Amiloides , Humanos , Análise Espectral Raman , Ressonância de Plasmônio de Superfície
19.
Int J Mol Sci ; 22(6)2021 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-33809300

RESUMO

Alzheimer's disease (AD), the most common form of neurodegenerative dementia in adults worldwide, is a multifactorial and heterogeneous disorder characterized by the interaction of genetic and epigenetic factors and the dysregulation of numerous intracellular signaling and cellular/molecular pathways. The introduction of the systems biology framework is revolutionizing the study of complex diseases by allowing the identification and integration of cellular/molecular pathways and networks of interaction. Here, we reviewed the relationship between physical activity and the next pathophysiological processes involved in the risk of developing AD, based on some crucial molecular pathways and biological process dysregulated in AD: (1) Immune system and inflammation; (2) Endothelial function and cerebrovascular insufficiency; (3) Apoptosis and cell death; (4) Intercellular communication; (5) Metabolism, oxidative stress and neurotoxicity; (6) DNA damage and repair; (7) Cytoskeleton and membrane proteins; (8) Synaptic plasticity. Moreover, we highlighted the increasingly relevant role played by advanced neuroimaging technologies, including structural/functional magnetic resonance imaging, diffusion tensor imaging, and arterial spin labelling, in exploring the link between AD and physical exercise. Regular physical exercise seems to have a protective effect against AD by inhibiting different pathophysiological molecular pathways implicated in AD.


Assuntos
Doença de Alzheimer/terapia , Exercício Físico/fisiologia , Estresse Oxidativo/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/reabilitação , Dano ao DNA/genética , Reparo do DNA/genética , Imagem de Tensor de Difusão/métodos , Humanos , Imageamento por Ressonância Magnética/métodos , Neuroimagem/métodos , Transdução de Sinais/genética
20.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34638786

RESUMO

Alzheimer's disease is a widespread and devastating neurological disorder associated with proteotoxic events caused by the misfolding and aggregation of the amyloid-ß peptide. To find therapeutic strategies to combat this disease, Drosophila melanogaster has proved to be an excellent model organism that is able to uncover anti-proteotoxic candidates due to its outstanding genetic toolbox and resemblance to human disease genes. In this review, we highlight the use of Drosophila melanogaster to both study the proteotoxicity of the amyloid-ß peptide and to screen for drug candidates. Expanding the knowledge of how the etiology of Alzheimer's disease is related to proteotoxicity and how drugs can be used to block disease progression will hopefully shed further light on the field in the search for disease-modifying treatments.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Doença de Alzheimer/genética , Peptídeos beta-Amiloides/genética , Animais , Modelos Animais de Doenças , Drosophila melanogaster , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa