Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; : 106694, 2024 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-39374707

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease and a leading cause of senile dementia. Amyloid-ß (Aß) accumulation triggers chronic neuroinflammation, initiating AD pathogenesis. Recent clinical trials for anti-Aß immunotherapy underscore that blood-based biomarkers have significant advantages and applicability over conventional diagnostics and are an unmet clinical need. To further advance ongoing clinical trials and identify novel therapeutic targets for AD, developing additional plasma biomarkers closely associated with pathogenic mechanisms downstream of Aß accumulation is critically important. To identify plasma metabolites reflective of neuroinflammation caused by Aß pathology, we performed untargeted metabolomic analyses of the plasma by capillary electrophoresis time-of-flight mass spectrometry (CE-TOFMS) and analyzed the potential roles of the identified metabolic changes in the brain neuroinflammatory response using the female App knock-in (AppNLGF) mouse model of Aß amyloidosis. The CE-TOFMS analysis of plasma samples from female wild-type (WT) and AppNLGF mice revealed that plasma levels of nicotinamide, a nicotinamide adenine dinucleotide (NAD+) precursor, were decreased in AppNLGF mice, and altered metabolite profiles were enriched for nicotinate/nicotinamide metabolism. In AppNLGF mouse brains, NAD+ levels were unaltered, but mRNA levels of NAD+-synthesizing nicotinate phosphoribosyltransferase (Naprt) and NAD+-degrading Cd38 genes were increased. These enzymes were induced in reactive astrocytes and microglia surrounding Aß plaques in the cortex and hippocampus of female AppNLGF mouse brains, suggesting neuroinflammation increases NAD+ metabolism. This study suggests plasma nicotinamide could be indicative of the neuroinflammatory response and that nicotinate and nicotinamide metabolism are potential therapeutic targets for AD, by targeting both neuroinflammation and neuroprotection.

2.
Chembiochem ; : e202400431, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39382238

RESUMEN

Alzheimer's disease (AD) is a multifactorial form of dementia mainly affecting people in the elderly, but no effective cure is available. According to the amyloid hypothesis the aggregation of Amyloid-ß (Aß) into oligomeric toxic species is believed to concur with the onset and progression of the disease heavily. By using a click chemistry approach, we conjugated a suitable designed peptide sequence to a metalloporphyrin moiety to obtain three hybrid peptide systems to be studied for their interact ion with Amyloid-ß peptides. The aim is to get new tools for the diagnosis and therapy in AD. The results described in this study, which were obtained through spectroscopic techniques (UV-Vis, CD, Bis-Ans and intrinsic porphyrin Fluorescence), Microfluidics (GCI) and cell biology (MTT, Live cell imaging and flow cytometry), reveal interesting features about the structure-activity relationships connecting these conjugates with the interaction with Aß, as well as on their potential use as sensing systems. In our opinion the data reported in this paper make the porphyrin-peptide conjugates highly compelling for further exploration as spectroscopic probes to detect Aß biomarkers in biological fluids.

3.
Mol Pharm ; 21(7): 3330-3342, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38875185

RESUMEN

The aberrant assembly of amyloid-ß (Aß) is implicated in Alzheimer's disease (AD). Recent clinical outcomes of Aß-targeted immunotherapy reinforce the notion that clearing Aß burden is a potential therapeutic approach for AD. Herein, to develop drug candidates for chemically driven clearance of Aß aggregates, we synthesized 51 novel polyfunctionalized furo[2,3-b:4,5-b']dipyridine-chalcone hybrid compounds. After conducting two types of cell-free anti-Aß functional assays, Aß aggregation prevention and Aß aggregate clearance, we selected YIAD-0336, (E)-8-((1H-pyrrol-2-yl)methylene)-10-(4-chlorophenyl)-2,4-dimethyl-7,8-dihydropyrido[3',2':4,5]furo[3,2-b]quinolin-9(6H)-one, for further in vivo investigations. As YIAD-0336 exhibited a low blood-brain barrier penetration profile, it was injected along with aggregated Aß directly into the intracerebroventricular region of ICR mice and ameliorated spatial memory in Y-maze tests. Next, YIAD-0336 was orally administered to 5XFAD transgenic mice with intravenous injections of mannitol, and YIAD-0336 significantly removed Aß plaques from the brains of 5XFAD mice. Collectively, YIAD-0336 dissociated toxic aggregates in the mouse brain and hence alleviated cognitive deterioration. Our findings indicate that chemically driven clearance of Aß aggregates is a promising therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Modelos Animales de Enfermedad , Ratones Transgénicos , Animales , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Ratones , Péptidos beta-Amiloides/metabolismo , Chalcona/química , Chalcona/farmacología , Chalcona/análogos & derivados , Chalconas/química , Chalconas/farmacología , Chalconas/administración & dosificación , Masculino , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Humanos , Memoria/efectos de los fármacos , Agregado de Proteínas/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Piridinas/química , Piridinas/farmacología , Piridinas/administración & dosificación
4.
Cereb Cortex ; 33(23): 11329-11338, 2023 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-37859548

RESUMEN

It is helpful to understand the pathology of Alzheimer's disease by exploring the relationship between amyloid-ß accumulation and cognition. The study explored the relationship between regional amyloid-ß accumulation and multiple cognitions and study their application value in the Alzheimer's disease diagnosis. 135 participants completed 18F-florbetapir Positron Emission Tomography (PET), structural MRI, and a cognitive battery. Partial correlation was used to examine the relationship between global and regional amyloid-ß accumulation and cognitions. Then, a support vector machine was applied to determine whether cognition-related accumulation regions can adequately distinguish the cognitively normal controls (76 participants) and mild cognitive impairment (30 participants) groups or mild cognitive impairment and Alzheimer's disease (29 participants) groups. The result showed that amyloid-ß accumulation regions were mainly located in the frontoparietal cortex, calcarine fissure, and surrounding cortex and temporal pole regions. Episodic memory-related regions included the frontoparietal cortices; executive function-related regions included the frontoparietal, temporal, and occipital cortices; and processing speed-related regions included the frontal and occipital cortices. Support vector machine analysis showed that only episodic memory-related amyloid-ß accumulation regions had better classification performance during the progression of Alzheimer's disease. Assessing regional changes in amyloid, particularly in frontoparietal regions, can aid in the early detection of amyloid-related decline in cognitive function.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Humanos , Anciano , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/patología , Disfunción Cognitiva/patología , Péptidos beta-Amiloides , Cognición , Tomografía de Emisión de Positrones/métodos , Amiloide
5.
Anal Biochem ; 661: 114996, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36427556

RESUMEN

Sleep debt (SD) is one of the important triggers for causing not only physiological and mental illness but also dangerous work. Therefore, achieving an early and objective assessment of SD is of great significance in the precaution against SD-related diseases and unsafe work. Here, an ultrasensitive electrochemical immunosensor was constructed for analysis of SD biomarker amyloid-ß (Aß). The gold nanoparticles/chitosan-coated polyaniline-functionalized activated carbon (AuNPs/AC@PANI@CS) composites were employed as the sensing platforms. Since PANI and AC can form an effective conductive path, it can effectively enhance the penetration of electrolytes on the electrode surface and the rapid transport of charges and ions, significantly enhancing the electrochemical response signal of the immunosensor. Under the optimized experimental conditions, the fabricated immunosensor had a wide linear range of 1.95 pg mL-1 to 1000.00 pg mL-1, with a low detection limit of 0.014 pg mL-1. This study not only provides an effective method for the accurate and rapid detection of Aß, but also offers a novel evaluation strategy for the objective assessment of SD and the study of related pathological mechanisms.


Asunto(s)
Técnicas Biosensibles , Nanopartículas del Metal , Humanos , Privación de Sueño , Oro , Inmunoensayo
6.
Angew Chem Int Ed Engl ; 62(7): e202210209, 2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36316282

RESUMEN

Amyloid-ß (Aß) in the form of neurotoxic aggregates is regarded as the main pathological initiator and key therapeutic target of Alzheimer's disease. However, anti-Aß drug development has been impeded by the lack of a target needed for structure-based drug design and low permeability of the blood-brain barrier (BBB). An attractive therapeutic strategy is the development of amyloid-based anti-Aß peptidomimetics that exploit the self-assembling nature of Aß and penetrate the BBB. Herein, we designed a dimeric peptide drug candidate based on the N-terminal fragment of Aß, DAB, found to cross the BBB and solubilize Aß oligomers and fibrils. Administration of DAB reduced amyloid burden in 5XFAD mice, and downregulated neuroinflammation and prevented memory impairment in the Y-maze test. Peptide mapping assays and molecular docking studies were utilized to elucidate DAB-Aß interaction. To further understand the active regions of DAB, we assessed the dissociative activity of DAB with sequence modifications.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Ratones , Animales , Simulación del Acoplamiento Molecular , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/química , Amiloide , Disfunción Cognitiva/tratamiento farmacológico , Ratones Transgénicos
7.
Neurobiol Dis ; 164: 105631, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35041991

RESUMEN

The APOE genotype is the most prominent genetic risk factor for the development of late-onset Alzheimer''s disease (LOAD); however, the underlying mechanisms remain unclear. In the present study, we found that the sialylation profiles of ApoE protein in the human brain are significantly different among the three isoforms, with ApoE2 exhibiting the most abundant sialic acid modification whereas ApoE4 had the least. We further observed that the sialic acid moiety in ApoE2 significantly affected the interaction between ApoE2 and Aß peptides. The removal of sialic acid in ApoE2 increased the ApoE2 binding affinity for the Aß17-24 region of Aß and promoted Aß fibrillation. These findings provide a plausible explanation for the well-documented differential roles of ApoE isoforms in Aß pathogenesis. Specifically, compared to the other two isotypes, the higher expression of sialic acid in ApoE2 may contribute to the less potent interaction between ApoE2 and Aß and ultimately the slower rate of brain Aß deposition, a mechanism thought to underlie ApoE2-mediated decreased risk for AD. Future studies are warranted to determine whether the differential sialylation in ApoE isoforms may also contribute to some of their other distinct properties, such as their divergent preferences in associations with lipids and lipoproteins, as well as their potential impact on neuroinflammation through modulation of microglial Siglec activity. Overall, our findings lead to the insight that the sialic acid structure is an important posttranslational modification (PTM) that alters ApoE protein functions with relevance for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Apolipoproteínas E/metabolismo , Isoformas de Proteínas/metabolismo , Encéfalo/metabolismo , Humanos , Ácido N-Acetilneuramínico/metabolismo
8.
J Biol Chem ; 295(30): 10138-10152, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32385113

RESUMEN

Mitochondrial dysfunction has long been implicated in the neurodegenerative disorder Parkinson's disease (PD); however, it is unclear how mitochondrial impairment and α-synuclein pathology are coupled. Using specific mitochondrial inhibitors, EM analysis, and biochemical assays, we report here that intramitochondrial protein homeostasis plays a major role in α-synuclein aggregation. We found that interference with intramitochondrial proteases, such as HtrA2 and Lon protease, and mitochondrial protein import significantly aggravates α-synuclein seeding. In contrast, direct inhibition of mitochondrial complex I, an increase in intracellular calcium concentration, or formation of reactive oxygen species, all of which have been associated with mitochondrial stress, did not affect α-synuclein pathology. We further demonstrate that similar mechanisms are involved in amyloid-ß 1-42 (Aß42) aggregation. Our results suggest that, in addition to other protein quality control pathways, such as the ubiquitin-proteasome system, mitochondria per se can influence protein homeostasis of cytosolic aggregation-prone proteins. We propose that approaches that seek to maintain mitochondrial fitness, rather than target downstream mitochondrial dysfunction, may aid in the search for therapeutic strategies to manage PD and related neuropathologies.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Mitocondrias/metabolismo , Enfermedad de Parkinson/metabolismo , Fragmentos de Péptidos/metabolismo , Proteostasis , alfa-Sinucleína/metabolismo , Péptidos beta-Amiloides/genética , Animales , Línea Celular Tumoral , Femenino , Serina Peptidasa A2 que Requiere Temperaturas Altas/genética , Serina Peptidasa A2 que Requiere Temperaturas Altas/metabolismo , Humanos , Mitocondrias/genética , Mitocondrias/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Fragmentos de Péptidos/genética , Ratas , Ratas Sprague-Dawley , Factores de Empalme Serina-Arginina/genética , Factores de Empalme Serina-Arginina/metabolismo , alfa-Sinucleína/genética
9.
FASEB J ; 34(10): 13272-13283, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32779267

RESUMEN

Transferrin receptor 1 (TfR1) mediated transcytosis is an attractive strategy to enhance brain uptake of protein drugs, but translation remains a challenge. Here, a single domain shark antibody VNAR fragment (TXB2) with similar affinity to murine and human TfR1 was used to shuttle protein cargo into the brain. TXB2 was fused to a human IgG1 Fc domain (hFc) or to the amyloid-ß (Aß) antibody bapineuzumab (Bapi). TXB2-hFc displayed 20-fold higher brain concentrations compared with a control VNAR-hFc at 18 hours post-injection in wt mice. At the same time point, brain concentrations of Bapi-TXB2 was threefold higher than Bapi. In transgenic mice overexpressing human Aß, the brain-to-blood concentration ratio increased with time due to interaction with intracerebral Aß deposits. The relatively stable threefold difference between Bapi-TXB2 and Bapi was observed for up to 6 days after injection. PET imaging and ex vivo autoradiography revealed more parenchymal distribution of Bapi-TXB2 compared with Bapi. In conclusion, the TXB2 VNAR shuttle markedly increased brain uptake of protein cargo and increased brain concentrations of the Aß binding antibody Bapi.


Asunto(s)
Antígenos CD/metabolismo , Productos Biológicos/administración & dosificación , Barrera Hematoencefálica/metabolismo , Receptores de Transferrina/metabolismo , Tromboxano B2/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/genética , Productos Biológicos/farmacocinética , Barrera Hematoencefálica/diagnóstico por imagen , Sistemas de Liberación de Medicamentos , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/genética , Tromboxano B2/genética , Transcitosis
10.
Int J Mol Sci ; 23(1)2021 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-35008544

RESUMEN

Endoplasmic reticulum (ER) degradation-enhancing α-mannosidase-like protein 1 (EDEM1) is a quality control factor directly involved in the endoplasmic reticulum-associated degradation (ERAD) process. It recognizes terminally misfolded proteins and directs them to retrotranslocation which is followed by proteasomal degradation in the cytosol. The amyloid-ß precursor protein (APP) is synthesized and N-glycosylated in the ER and transported to the Golgi for maturation before being delivered to the cell surface. The amyloidogenic cleavage pathway of APP leads to production of amyloid-ß (Aß), deposited in the brains of Alzheimer's disease (AD) patients. Here, using biochemical methods applied to human embryonic kidney, HEK293, and SH-SY5Y neuroblastoma cells, we show that EDEM1 is an important regulatory factor involved in APP metabolism. We find that APP cellular levels are significantly reduced after EDEM1 overproduction and are increased in cells with downregulated EDEM1. We also report on EDEM1-dependent transport of APP from the ER to the cytosol that leads to proteasomal degradation of APP. EDEM1 directly interacts with APP. Furthermore, overproduction of EDEM1 results in decreased Aß40 and Aß42 secretion. These findings indicate that EDEM1 is a novel regulator of APP metabolism through ERAD.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de la Membrana/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo , Línea Celular , Línea Celular Tumoral , Citosol/metabolismo , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico/fisiología , Glicosilación , Aparato de Golgi/metabolismo , Células HEK293 , Humanos , Pliegue de Proteína , alfa-Manosidasa/metabolismo
11.
Molecules ; 26(10)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064783

RESUMEN

All over the world, societies are facing rapidly aging populations combined with a growing number of patients suffering from Alzheimer's disease (AD). One focus in pharmaceutical research to address this issue is on the reduction of the longer amyloid-ß (Aß) fragments in the brain by modulation of γ-secretase, a membrane-bound protease. R-Flurbiprofen (tarenflurbil) was studied in this regard but failed to show significant improvement in AD patients in a phase 3 clinical trial. This was mainly attributed to its low ability to cross the blood-brain barrier (BBB). Here, we present the synthesis and in vitro evaluation of a racemic meta-carborane analogue of flurbiprofen. By introducing the carborane moiety, the hydrophobicity could be shifted into a more favourable range for the penetration of the blood-brain barrier, evident by a logD7.4 value of 2.0. Furthermore, our analogue retained γ-secretase modulator activity in comparison to racemic flurbiprofen in a cell-based assay. These findings demonstrate the potential of carboranes as phenyl mimetics also in AD research.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Compuestos de Boro/farmacología , Flurbiprofeno/análogos & derivados , Compuestos de Boro/síntesis química , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores de la Ciclooxigenasa/farmacología , Flurbiprofeno/química , Humanos , Concentración 50 Inhibidora
12.
J Biol Chem ; 294(21): 8438-8451, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-30918024

RESUMEN

Antibodies that recognize amyloidogenic aggregates with high conformational and sequence specificity are important for detecting and potentially treating a wide range of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. However, these types of antibodies are challenging to generate because of the large size, hydrophobicity, and heterogeneity of protein aggregates. To address this challenge, we developed a method for generating antibodies specific for amyloid aggregates. First, we grafted amyloidogenic peptide segments from the target polypeptide [Alzheimer's amyloid-ß (Aß) peptide] into the complementarity-determining regions (CDRs) of a stable antibody scaffold. Next, we diversified the grafted and neighboring CDR sites using focused mutagenesis to sample each WT or grafted residue, as well as one to five of the most commonly occurring amino acids at each site in human antibodies. Finally, we displayed these antibody libraries on the surface of yeast cells and selected antibodies that strongly recognize Aß-amyloid fibrils and only weakly recognize soluble Aß. We found that this approach enables the generation of monovalent and bivalent antibodies with nanomolar affinity for Aß fibrils. These antibodies display high conformational and sequence specificity as well as low levels of nonspecific binding and recognize a conformational epitope at the extreme N terminus of human Aß. We expect that this systematic approach will be useful for generating antibodies with conformational and sequence specificity against a wide range of peptide and protein aggregates associated with neurodegenerative disorders.


Asunto(s)
Péptidos beta-Amiloides , Regiones Determinantes de Complementariedad , Anticuerpos de Cadena Única , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/inmunología , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/inmunología , Humanos , Mutagénesis Sitio-Dirigida , Anticuerpos de Cadena Única/química , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología
13.
J Biol Chem ; 294(22): 8806-8818, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-30996005

RESUMEN

Aß1-42 is involved in Alzheimer's disease (AD) pathogenesis and is prone to glycation, an irreversible process where proteins accumulate advanced glycated end products (AGEs). Nϵ-(Carboxyethyl)lysine (CEL) is a common AGE associated with AD patients and occurs at either Lys-16 or Lys-28 of Aß1-42. Methyglyoxal is commonly used for the unspecific glycation of Aß1-42, which results in a complex mixture of AGE-modified peptides and makes interpretation of a causative AGE at a specific amino acid residue difficult. We address this issue by chemically synthesizing defined CEL modifications on Aß1-42 at Lys-16 (Aß-CEL16), Lys-28 (Aß-CEL28), and Lys-16 and -28 (Aß-CEL16&28). We demonstrated that double-CEL glycations at Lys-16 and Lys-28 of Aß1-42 had the most profound impact on the ability to form amyloid fibrils. In silico predictions indicated that Aß-CEL16&28 had a substantial decrease in free energy change, which contributes to fibril destabilization, and a increased aggregation rate. Single-CEL glycations at Lys-28 of Aß1-42 had the least impact on fibril formation, whereas CEL glycations at Lys-16 of Aß1-42 delayed fibril formation. We also tested these peptides for neuronal toxicity and mitochondrial function on a retinoic acid-differentiated SH-SY5Y human neuroblastoma cell line (RA-differentiated SH-SY5Y). Only Aß-CEL16 and Aß-CEL28 were neurotoxic, possibly through a nonmitochondrial pathway, whereas Aß-CEL16&28 showed no neurotoxicity. Interestingly, Aß-CEL16&28 had depolarized the mitochondrial membrane potential, whereas Aß-CEL16 had increased mitochondrial respiration at complex II. These results may indicate mitophagy or an alternate route of metabolism, respectively. Therefore, our results provides insight into potential therapeutic approaches against neurotoxic CEL-glycated Aß1-42.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/síntesis química , Péptidos beta-Amiloides/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Glicosilación , Humanos , Lisina/análogos & derivados , Lisina/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/toxicidad , Agregado de Proteínas , Conformación Proteica en Lámina beta , Estabilidad Proteica , Oxígeno Singlete/metabolismo
14.
J Biol Chem ; 294(5): 1478-1487, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30504224

RESUMEN

The 42-amino-acid ß-amyloid (Aß42) is a critical causative agent in the pathology of Alzheimer's disease. The hereditary Arctic mutation of Aß42 (E22G) leads to increased intracellular accumulation of ß-amyloid in early-onset Alzheimer's disease. However, it remains largely unknown how the Arctic mutant variant leads to aggressive protein aggregation and increased intracellular toxicity. Here, we constructed stable cell lines expressing fluorescent-tagged wildtype (WT) and E22G Aß42 to study the aggregation kinetics of the Arctic Aß42 mutant peptide and its heterogeneous structural forms. Arctic-mutant peptides assemble and form fibrils at a much faster rate than WT peptides. We identified five categories of intracellular aggregate-oligomers, single fibrils, fibril bundles, clusters, and aggresomes-that underline the heterogeneity of these Aß42 aggregates and represent the progression of Aß42 aggregation within the cell. Fluorescence-lifetime imaging (FLIM) and 3D structural illumination microscopy (SIM) showed that all aggregate species displayed highly compact structures with strong affinity between individual fibrils. We also found that aggregates formed by Arctic mutant Aß42 were more resistant to intracellular degradation than their WT counterparts. Our findings uncover the structural basis of the progression of Arctic mutant Aß42 aggregation in the cell.


Asunto(s)
Péptidos beta-Amiloides/química , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Mutación , Imagen Óptica/métodos , Multimerización de Proteína , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/ultraestructura , Humanos , Cinética , Modelos Moleculares , Conformación Proteica
15.
J Biol Chem ; 294(15): 6042-6053, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30787106

RESUMEN

Oligomeric assemblies of amyloid-ß (Aß) peptide (Aßo) in the brains of individuals with Alzheimer's disease (AD) are toxic to neuronal synapses. More than a dozen Aß receptor candidates have been suggested to be responsible for various aspects of the molecular pathology and memory impairment in mouse models of AD. A lack of consistent experimental design among previous studies of different receptor candidates limits evaluation of the relative roles of these candidates, producing some controversy within the field. Here, using cell-based assays with several Aß species, including Aßo from AD brains obtained by autopsy, we directly compared the Aß-binding capacity of multiple receptor candidates while accounting for variation in expression and confirming cell surface expression. In a survey of 15 reported Aß receptors, only cellular prion protein (PrPC), Nogo receptor 1 (NgR1), and leukocyte immunoglobulin-like receptor subfamily B member 2 (LilrB2) exhibited direct binding to synaptotoxic assemblies of synthetic Aß. Both PrPC and NgR1 preferentially bound synaptotoxic oligomers rather than nontoxic monomers, and the method of oligomer preparation did not significantly alter our binding results. Hippocampal neurons lacking both NgR1 and LilrB2 exhibited a partial reduction of Aßo binding, but this reduction was lower than in neurons lacking PrPC under the same conditions. Finally, binding studies with soluble Aßo from human AD brains revealed a strong affinity for PrPC, weak affinity for NgR1, and no detectable affinity for LilrB2. These findings clarify the relative contributions of previously reported Aß receptors under controlled conditions and highlight the prominence of PrPC as an Aß-binding site.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptor Nogo 1/metabolismo , Proteínas PrPC/metabolismo , Receptores Inmunológicos/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuronas/patología , Receptor Nogo 1/genética , Proteínas PrPC/genética , Receptores Inmunológicos/genética
16.
BMC Med Genet ; 21(1): 181, 2020 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-32919460

RESUMEN

BACKGROUND: The complement component (3b/4b) receptor 1 gene (CR1) gene has been proved to affect the susceptibility of Alzheimer's disease (AD) in different ethnic and districts groups. However, the effect of CR1 genetic variants on amyloid ß (Aß) metabolism of AD human is still unclear. Hence, the aim of this study was to investigate genetic influences of CR1 gene on Aß metabolism. METHODS: All data of AD patients and normal controls (NC) were obtained from alzheimer's disease neuroimaging initiative database (ADNI) database. In order to assess the effect of each single nucleotide polymorphism (SNP) of CR1 on Aß metabolism, the PLINK software was used to conduct the quality control procedures to enroll appropriate SNPs. Moreover, the correlation between CR1 genotypes and Aß metabolism in all participants were estimated with multiple linear regression models. RESULTS: After quality control procedures, a total of 329 samples and 83 SNPs were enrolled in our study. Moreover, our results identified five SNPs (rs10494884, rs11118322, rs1323721, rs17259045 and rs41308433), which were linked to Aß accumulation in brain. In further analyses, rs17259045 was found to decrease Aß accumulation among AD patients. Additionally, our study revealed the genetic variants in rs12567945 could increase CSF Aß42 in NC population. CONCLUSIONS: Our study had revealed several novel SNPs in CR1 genes which might be involved in the progression of AD via regulating Aß accumulation. These findings will provide a new basis for the diagnosis and treatment AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/líquido cefalorraquídeo , Biomarcadores/líquido cefalorraquídeo , Neuroimagen/métodos , Fragmentos de Péptidos/líquido cefalorraquídeo , Polimorfismo de Nucleótido Simple , Receptores de Complemento 3b/genética , Anciano , Anciano de 80 o más Años , Alelos , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Biomarcadores/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Femenino , Genotipo , Humanos , Masculino , Fragmentos de Péptidos/metabolismo
17.
J Biol Chem ; 293(40): 15419-15428, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30143530

RESUMEN

The histopathology of Alzheimer's disease (AD) is characterized by neuronal loss, neurofibrillary tangles, and senile plaque formation. The latter results from an exacerbated production (familial AD cases) or altered degradation (sporadic cases) of 40/42-amino acid-long ß-amyloid peptides (Aß peptides) that are produced by sequential cleavages of Aß precursor protein (ßAPP) by ß- and γ-secretases. The amyloid cascade hypothesis proposes a key role for the full-length Aß42 and the Aß40/42 ratio in AD etiology, in which soluble Aß oligomers lead to neurotoxicity, tau hyperphosphorylation, aggregation, and, ultimately, cognitive defects. However, following this postulate, during the last decade, several clinical approaches aimed at decreasing full-length Aß42 production or neutralizing it by immunotherapy have failed to reduce or even stabilize AD-related decline. Thus, the Aß peptide (Aß40/42)-centric hypothesis is probably a simplified view of a much more complex situation involving a multiplicity of APP fragments and Aß catabolites. Indeed, biochemical analyses of AD brain deposits and fluids have unraveled an Aß peptidome consisting of additional Aß-related species. Such Aß catabolites could be due to either primary enzymatic cleavages of ßAPP or secondary processing of Aß itself by exopeptidases. Here, we review the diversity of N- and C-terminally truncated Aß peptides and their biosynthesis and outline their potential function/toxicity. We also highlight their potential as new pharmaceutical targets and biomarkers.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Región CA1 Hipocampal/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/genética , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animales , Anticuerpos Monoclonales/farmacología , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Nootrópicos/farmacología , Dominios Proteicos , Proteolisis , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo
18.
J Biol Chem ; 293(9): 3118-3125, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29298895

RESUMEN

Endoplasmic reticulum (ER) stress is believed to play an important role in the etiology of Alzheimer's disease (AD). The accumulation of misfolded proteins and perturbation of intracellular calcium homeostasis are thought to underlie the induction of ER stress, resulting in neuronal dysfunction and cell death. Several reports have shown an increased ER stress response in amyloid precursor protein (APP) and presenilin1 (PS1) double-transgenic (Tg) AD mouse models. However, whether the ER stress observed in these mouse models is actually caused by AD pathology remains unclear. APP and PS1 contain one and nine transmembrane domains, respectively, for which it has been postulated that overexpressed membrane proteins can become wedged in a misfolded configuration in ER membranes, thereby inducing nonspecific ER stress. Here, we used an App-knockin (KI) AD mouse model that accumulates amyloid-ß (Aß) peptide without overexpressing APP to investigate whether the ER stress response is heightened because of Aß pathology. Thorough examinations indicated that no ER stress responses arose in App-KI or single APP-Tg mice. These results suggest that PS1 overexpression or mutation induced a nonspecific ER stress response that was independent of Aß pathology in the double-Tg mice. Moreover, we observed no ER stress in a mouse model of tauopathy (P301S-Tau-Tg mice) at various ages, suggesting that ER stress is also not essential in tau pathology-induced neurodegeneration. We conclude that the role of ER stress in AD pathogenesis needs to be carefully addressed in future studies.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Técnicas de Sustitución del Gen , Animales , Línea Celular , Expresión Génica , Ratones , Ratones Endogámicos C57BL
19.
J Biol Chem ; 293(1): 226-244, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29127203

RESUMEN

Patients recovering from sepsis have higher rates of CNS morbidities associated with long-lasting impairment of cognitive functions, including neurodegenerative diseases. However, the molecular etiology of these sepsis-induced impairments is unclear. Here, we investigated the role of the receptor for advanced glycation end products (RAGE) in neuroinflammation, neurodegeneration-associated changes, and cognitive dysfunction arising after sepsis recovery. Adult Wistar rats underwent cecal ligation and perforation (CLP), and serum and brain (hippocampus and prefrontal cortex) samples were obtained at days 1, 15, and 30 after the CLP. We examined these samples for systemic and brain inflammation; amyloid-ß peptide (Aß) and Ser-202-phosphorylated Tau (p-TauSer-202) levels; and RAGE, RAGE ligands, and RAGE intracellular signaling. Serum markers associated with the acute proinflammatory phase of sepsis (TNFα, IL-1ß, and IL-6) rapidly increased and then progressively decreased during the 30-day period post-CLP, concomitant with a progressive increase in RAGE ligands (S100B, Nϵ-[carboxymethyl]lysine, HSP70, and HMGB1). In the brain, levels of RAGE and Toll-like receptor 4, glial fibrillary acidic protein and neuronal nitric-oxide synthase, and Aß and p-TauSer-202 also increased during that time. Of note, intracerebral injection of RAGE antibody into the hippocampus at days 15, 17, and 19 post-CLP reduced Aß and p-TauSer-202 accumulation, Akt/mechanistic target of rapamycin signaling, levels of ionized calcium-binding adapter molecule 1 and glial fibrillary acidic protein, and behavioral deficits associated with cognitive decline. These results indicate that brain RAGE is an essential factor in the pathogenesis of neurological disorders following acute systemic inflammation.


Asunto(s)
Productos Finales de Glicación Avanzada/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Cognición/fisiología , Disfunción Cognitiva/metabolismo , Hipocampo/metabolismo , Inflamación/metabolismo , Masculino , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Fosforilación , Ratas , Ratas Wistar , Sepsis/complicaciones , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas tau/metabolismo
20.
J Biol Chem ; 293(8): 2888-2902, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29282295

RESUMEN

Amyloid-ß (Aß) and human islet amyloid polypeptide (hIAPP) aggregate to form amyloid fibrils that deposit in tissues and are associated with Alzheimer's disease (AD) and type II diabetes (T2D), respectively. Individuals with T2D have an increased risk of developing AD, and conversely, AD patients have an increased risk of developing T2D. Evidence suggests that this link between AD and T2D might originate from a structural similarity between aggregates of Aß and hIAPP. Using the cryoEM method microelectron diffraction, we determined the atomic structures of 11-residue segments from both Aß and hIAPP, termed Aß(24-34) WT and hIAPP(19-29) S20G, with 64% sequence similarity. We observed a high degree of structural similarity between their backbone atoms (0.96-Å root mean square deviation). Moreover, fibrils of these segments induced amyloid formation through self- and cross-seeding. Furthermore, inhibitors designed for one segment showed cross-efficacy for full-length Aß and hIAPP and reduced cytotoxicity of both proteins, although by apparently blocking different cytotoxic mechanisms. The similarity of the atomic structures of Aß(24-34) WT and hIAPP(19-29) S20G offers a molecular model for cross-seeding between Aß and hIAPP.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Modelos Moleculares , Ovillos Neurofibrilares/metabolismo , Fragmentos de Péptidos/metabolismo , Sustitución de Aminoácidos , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/genética , Animales , Línea Celular Tumoral , Biología Computacional , Cristalografía por Rayos X , Diseño de Fármacos , Células HEK293 , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/ultraestructura , Polipéptido Amiloide de los Islotes Pancreáticos/antagonistas & inhibidores , Polipéptido Amiloide de los Islotes Pancreáticos/química , Polipéptido Amiloide de los Islotes Pancreáticos/genética , Ratones , Microscopía Electrónica de Transmisión , Mutación , Ovillos Neurofibrilares/química , Ovillos Neurofibrilares/efectos de los fármacos , Ovillos Neurofibrilares/ultraestructura , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Neuronas/ultraestructura , Nootrópicos/química , Nootrópicos/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Agregación Patológica de Proteínas/metabolismo , Agregación Patológica de Proteínas/patología , Agregación Patológica de Proteínas/prevención & control , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA