Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biomacromolecules ; 25(6): 3583-3595, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38703359

RESUMEN

Polyglycidol or polyglycerol (PG), a polyether widely used in biomedical applications, has not been extensively studied in its branched cyclic form (bcPG), despite extensive research on hyperbranched PG (HPG). This study explores the biomedical promise of bcPG, particularly its ability to cross the blood-brain barrier (BBB). We evaluate in vitro biocompatibility, endothelial permeability, and formation of branched linear PG (blPG) as topological impurities in the presence of water. Small angle X-ray scattering in solution revealed a fractal dimension of approximately two for bcPG and the mixture bc+blPG, suggesting random branching. Comparisons of cytotoxicity and endothelial permeability between bcPG, bc+blPG, and HPG in a BBB model using hCMEC/D3 cells showed different biocompatibility profiles and higher endothelial permeability for HPG. bcPG showed a tendency to accumulate around cell nuclei, in contrast to the behavior of HPG. This study contributes to the understanding of the influence of polymer topology on biological behavior.


Asunto(s)
Materiales Biocompatibles , Barrera Hematoencefálica , Humanos , Barrera Hematoencefálica/metabolismo , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Polimerizacion , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Polímeros/química , Polímeros/farmacología , Glicerol/química , Compuestos Epoxi/química , Línea Celular , Permeabilidad , Glicoles de Propileno/química , Propanoles/química
2.
Analyst ; 148(23): 6070-6077, 2023 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-37904570

RESUMEN

The application of liposomes (LPs) to central nervous system disorders could represents a turning point in the therapy and quality of life of patients. Indeed, LPs have demonstrated their ability to cross the blood-brain barrier (BBB) and, as a consequence, to enhance the therapeutics delivery into the brain. Some approaches for BBB crossing involve the modification of LP surfaces with biologically active ligands. Among them, the Apolipoprotein E-modified peptide (mApoE) has been used for several LP-based nanovectors under investigation. In this study, we propose Surface Plasmon Resonance imaging (SPRi) for the characterization of multifunctionalized LPs for Glioblastoma treatment. LPs were functionalized with mApoE and with a metallo-protease sensitive lipopeptide to deliver and guarantee the localized release of an encapsulated drug in diseased areas. The SPRi analysis was optimized in order to evaluate the binding affinity between LPs and mApoE receptors, finding that mApoE-LPs generated SPRi signals referred to interactions between mApoE and receptors mainly present in the brain. Moreover, a significant binding between LPs and VCAM-1 (endothelial receptor) was observed, whereas LPs did not interact significantly with peripheral receptors expressed on monocytes and lymphocytes. SPRi results confirmed not only the presence of mApoE on LP surfaces, but also its binding affinity, thanks to the specific interaction with selected receptors. In conclusion, the high sensitivity and the multiplexing capability associated with the low volumes of sample required and the minimal sample preparation, make SPRi an excellent technique for the characterization of multifunctionalized nanoparticles-based formulations.


Asunto(s)
Encefalopatías , Liposomas , Humanos , Lipopolisacáridos , Calidad de Vida , Resonancia por Plasmón de Superficie , Sistemas de Liberación de Medicamentos
3.
J Nanobiotechnology ; 17(1): 115, 2019 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-31711496
4.
Int J Mol Sci ; 20(7)2019 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-30970594

RESUMEN

The cell microenvironment plays a pivotal role in mediating cell adhesion, survival, and proliferation in physiological and pathological states. The relevance of extracellular matrix (ECM) proteins in cell fate control is an important issue to take into consideration for both tissue engineering and cell biology studies. The glycosylation of ECM proteins remains, however, largely unexplored. In order to investigate the physio-pathological effects of differential ECM glycosylation, the design of affordable chemoselective methods for ECM components glycosylation is desirable. We will describe a new chemoselective glycosylation approach exploitable in aqueous media and on non-protected substrates, allowing rapid access to glyco-functionalized biomaterials.


Asunto(s)
Materiales Biocompatibles/metabolismo , Técnicas de Cultivo de Célula/métodos , Proteínas de la Matriz Extracelular/metabolismo , Materiales Biocompatibles/química , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Colágeno/química , Colágeno/farmacología , Glicosilación , Humanos
5.
Nanomedicine ; 14(2): 609-618, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29248676

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder related, in part, to the accumulation of amyloid-ß peptide (Aß) and especially the Aß peptide 1-42 (Aß1-42). The aim of this study was to design nanocarriers able to: (i) interact with the Aß1-42 in the blood and promote its elimination through the "sink effect" and (ii) correct the memory defect observed in AD-like transgenic mice. To do so, biodegradable, PEGylated nanoparticles were surface-functionalized with an antibody directed against Aß1-42. Treatment of AD-like transgenic mice with anti-Aß1-42-functionalized nanoparticles led to: (i) complete correction of the memory defect; (ii) significant reduction of the Aß soluble peptide and its oligomer level in the brain and (iii) significant increase of the Aß levels in plasma. This study represents the first example of Aß1-42 monoclonal antibody-decorated nanoparticle-based therapy against AD leading to complete correction of the memory defect in an experimental model of AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales/química , Modelos Animales de Enfermedad , Trastornos de la Memoria/terapia , Nanopartículas/administración & dosificación , Polímeros/administración & dosificación , Animales , Anticuerpos Monoclonales/inmunología , Humanos , Masculino , Ratones , Ratones Transgénicos , Nanopartículas/química , Nanopartículas/metabolismo , Polímeros/química , Polímeros/metabolismo , Recuperación de la Función
6.
Biochim Biophys Acta ; 1860(4): 746-56, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26774643

RESUMEN

BACKGROUND: Amyloid ß (Aß) peptide aggregation is the main molecular mechanism underlying the development of Alzheimer's disease, the most widespread form of senile dementia worldwide. Increasing evidence suggests that the key factor leading to impaired neuronal function is accumulation of water-soluble Aß oligomers rather than formation of the senile plaques created by the deposition of large fibrillary aggregates of Aß. However, several questions remain about the preliminary steps and the progression of Aß oligomerization. METHODS: We show that the initial stages of the aggregation of fluorescently labeled Aß can be determined with a high degree of precision and at physiological (i.e., nanomolar) concentrations by using either steady-state fluorimetry or time-correlated single-photon counting. RESULTS: We study the dependence of the oligomerization extent and rate on the Aß concentration. We determine the chemical binding affinity of fluorescently labeled Aß for liposomes that have been recently shown to be pharmacologically active in vivo, reducing the Aß burden within the brain. We also probe their capacity to hinder the Aß oligomerization process in vitro. CONCLUSIONS: We introduced a fluorescence assay allowing investigation of the earliest steps of Aß oligomerization, the peptide involved in Alzheimer's disease. The assay proved to be sensitive even at Aß concentrations as low as those physiologically observed in the cerebrospinal fluid. GENERAL SIGNIFICANCE: This work represents an extensive and quantitative study on the initial events of Aß oligomerization at physiological concentration. It may enhance our comprehension of the molecular mechanisms leading to Alzheimer's disease, thus paving the way to novel therapeutic strategies.


Asunto(s)
Péptidos beta-Amiloides/química , Liposomas/química , Fragmentos de Péptidos/química , Agregación Patológica de Proteínas , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Humanos , Fragmentos de Péptidos/metabolismo , Espectrometría de Fluorescencia
7.
Nanomedicine ; 13(2): 723-732, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27769888

RESUMEN

Aggregation of amyloid-ß peptide (Aß) is a key event in the pathogenesis of Alzheimer's disease (AD). We investigated the effects of nanoliposomes decorated with the retro-inverso peptide RI-OR2-TAT (Ac-rGffvlkGrrrrqrrkkrGy-NH2) on the aggregation and toxicity of Aß. Remarkably low concentrations of these peptide inhibitor nanoparticles (PINPs) were required to inhibit the formation of Aß oligomers and fibrils in vitro, with 50% inhibition occurring at a molar ratio of ~1:2000 of liposome-bound RI-OR2-TAT to Aß. PINPs also bound to Aß with high affinity (Kd=13.2-50 nM), rescued SHSY-5Y cells from the toxic effect of pre-aggregated Aß, crossed an in vitro blood-brain barrier model (hCMEC/D3 cell monolayer), entered the brains of C57 BL/6 mice, and protected against memory loss in APPSWE transgenic mice in a novel object recognition test. As the most potent aggregation inhibitor that we have tested so far, we propose to develop PINPs as a potential disease-modifying treatment for AD.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/terapia , Nanopartículas , Fragmentos de Péptidos , Péptidos beta-Amiloides , Animales , Barrera Hematoencefálica , Humanos , Liposomas , Ratones Transgénicos , Células Tumorales Cultivadas
8.
J Neurosci ; 34(42): 14022-31, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25319699

RESUMEN

Alzheimer's disease is characterized by the accumulation and deposition of plaques of ß-amyloid (Aß) peptide in the brain. Given its pivotal role, new therapies targeting Aß are in demand. We rationally designed liposomes targeting the brain and promoting the disaggregation of Aß assemblies and evaluated their efficiency in reducing the Aß burden in Alzheimer's disease mouse models. Liposomes were bifunctionalized with a peptide derived from the apolipoprotein-E receptor-binding domain for blood-brain barrier targeting and with phosphatidic acid for Aß binding. Bifunctionalized liposomes display the unique ability to hinder the formation of, and disaggregate, Aß assemblies in vitro (EM experiments). Administration of bifunctionalized liposomes to APP/presenilin 1 transgenic mice (aged 10 months) for 3 weeks (three injections per week) decreased total brain-insoluble Aß1-42 (-33%), assessed by ELISA, and the number and total area of plaques (-34%) detected histologically. Also, brain Aß oligomers were reduced (-70.5%), as assessed by SDS-PAGE. Plaque reduction was confirmed in APP23 transgenic mice (aged 15 months) either histologically or by PET imaging with [(11)C]Pittsburgh compound B (PIB). The reduction of brain Aß was associated with its increase in liver (+18%) and spleen (+20%). Notably, the novel-object recognition test showed that the treatment ameliorated mouse impaired memory. Finally, liposomes reached the brain in an intact form, as determined by confocal microscopy experiments with fluorescently labeled liposomes. These data suggest that bifunctionalized liposomes destabilize brain Aß aggregates and promote peptide removal across the blood-brain barrier and its peripheral clearance. This all-in-one multitask therapeutic device can be considered as a candidate for the treatment of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Apolipoproteínas E/administración & dosificación , Modelos Animales de Enfermedad , Liposomas/administración & dosificación , Trastornos de la Memoria/tratamiento farmacológico , Placa Amiloide/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteínas E/metabolismo , Liposomas/metabolismo , Masculino , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patología , Distribución Aleatoria
9.
Nanomedicine ; 11(2): 421-30, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25461285

RESUMEN

The accumulation of extracellular amyloid-beta (Aß) peptide and intracellular neurofibrillary tangles in the brain are two major neuropathological hallmarks of Alzheimer's disease (AD). It is thought that an equilibrium exists between Aß in the brain and in the peripheral blood and thus, it was hypothesized that shifting this equilibrium towards the blood by enhancing peripheral clearance might reduce Aß levels in the brain: the 'sink effect'. We tested this hypothesis by intraperitoneally injecting APP/PS1 transgenic mice with small unilamellar vesicles containing either phosphatidic acid or cardiolipin over 3weeks. This treatment reduced significantly the amount of Aß in the plasma and the brain levels of Aß were lighter affected. Nevertheless, this dosing regimen did modulate tau phosphorylation and glycogen synthase kinase 3 activities in the brain, suggesting that the targeting of circulating Aß may be therapeutically relevant in AD. FROM THE CLINICAL EDITOR: Intraperitoneal injection of small unilamellar vesicles containing phosphatidic acid or cardiolipin significantly reduced the amount of amyloid-beta (Aß) peptide in the plasma in a rodent model. Brain levels of Aß were also affected - although to a lesser extent - suggesting that targeting of circulating Aß may be therapeutically relevant of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/sangre , Cardiolipinas/administración & dosificación , Ácidos Fosfatidicos/administración & dosificación , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Cardiolipinas/química , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Inyecciones Intraperitoneales , Liposomas/administración & dosificación , Liposomas/química , Ratones , Ratones Transgénicos , Nanopartículas/administración & dosificación , Nanopartículas/química , Ácidos Fosfatidicos/química , Proteínas tau/metabolismo
10.
Nanomedicine ; 10(7): 1583-90, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24333591

RESUMEN

Targeting amyloid-ß peptide (Aß) within the brain is a strategy actively sought for therapy of Alzheimer's disease (AD). We investigated the ability of liposomes bi-functionalized with phosphatidic acid and with a modified ApoE-derived peptide (mApoE-PA-LIP) to affect Aß aggregation/disaggregation features and to cross in vitro and in vivo the blood-brain barrier (BBB). Surface plasmon resonance showed that bi-functionalized liposomes strongly bind Aß (kD=0.6 µM), while Thioflavin-T and SDS-PAGE/WB assays show that liposomes inhibit peptide aggregation (70% inhibition after 72 h) and trigger the disaggregation of preformed aggregates (60% decrease after 120 h incubation). Moreover, experiments with dually radiolabelled LIP suggest that bi-functionalization enhances the passage of radioactivity across the BBB either in vitro (permeability=2.5×10(-5) cm/min, 5-fold higher with respect to mono-functionalized liposomes) or in vivo in healthy mice. Taken together, our results suggest that mApoE-PA-LIP are valuable nanodevices with a potential applicability in vivo for the treatment of AD. From the clinical editor: Bi-functionalized liposomes with phosphatidic acid and a modified ApoE-derived peptide were demonstrated to influence Aß aggregation/disaggregation as a potential treatment in an Alzheimer's model. The liposomes were able to cross the blood-brain barrier in vitro and in vivo. Similar liposomes may become clinically valuable nanodevices with a potential applicability for the treatment of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/terapia , Apolipoproteínas E/química , Barrera Hematoencefálica , Liposomas , Péptidos/química , Ácidos Fosfatidicos/química , Apolipoproteínas E/administración & dosificación , Western Blotting , Electroforesis en Gel de Poliacrilamida , Humanos , Ácidos Fosfatidicos/administración & dosificación , Resonancia por Plasmón de Superficie
11.
Ann Med ; 55(1): 2205659, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37143345

RESUMEN

AIM: The effect of liposomes bi-functionalized with phosphatidic acid and with a synthetic peptide derived from human apolipoprotein E has been evaluated on the aggregation features of different amyloidogenic proteins: human Amyloid ß1-40 (Aß1-40), transthyretin (TTR) variant S52P, human ß2microglobulin (ß2m) variants ΔN6 and D76N, Serum Amyloid A (SAA). METHODS: The formation of fibrillar aggregates of the proteins was investigated by ThioflavinT fluorescence assay and validated by Atomic Force Microscopy. RESULTS: The results show that liposomes are preventing the transition of non-aggregated forms to the fibrillar state, with stronger effects on Aß1-40, ß2m ΔN6 and SAA. Liposomes also induce disaggregation of the amyloid aggregates of all the proteins investigated, with stronger effects on Aß1-40, ß2 D76N and TTR.SPR assays show that liposomes bind Aß1-40 and SAA aggregates with high affinity (KD in the nanomolar range) whereas binding to TTR aggregates showed a lower affinity (KD in the micromolar range). Aggregates of ß2m variants showed both high and low affinity binding sites. Computed Structural analysis of protein fibrillar aggregates and considerations on the multidentate features of liposomes allow to speculate a common mechanism of action, based on binding the ß-stranded peptide regions responsible for the amyloid formation. CONCLUSION: Thus, multifunctional liposomes perform as pharmacological chaperones with anti-amyloidogenic activity, with a promising potential for the treatment of a number of protein-misfolding diseases.Key messageAmyloidosis is a group of diseases, each due to a specific protein misfolding.Anti-amyloidogenic nanoparticles have been gaining the utmost importance as a potential treatment for protein misfolding disorders.Liposomes bi-functionalized with phosphatidic acid and with a synthetic peptide derived from human apolipoprotein E showed anti-amyloidogenic activity.


Asunto(s)
Amiloide , Liposomas , Humanos , Amiloide/química , Amiloide/metabolismo , Agregado de Proteínas , Chaperonas Moleculares , Ácidos Fosfatidicos , Apolipoproteínas
12.
Biochim Biophys Acta Biomembr ; 1864(1): 183763, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34506799

RESUMEN

Doxorubicin (DOX) is one of the most efficient antitumor drugs employed in numerous cancer therapies. Its incorporation into lipid-based nanocarriers, such as liposomes, improves the drug targeting into tumor cells and reduces drug side effects. The carriers' lipid composition is expected to affect the interactions of DOX and its partitioning into liposomal membranes. To get a rational insight into this aspect and determine promising lipid compositions, we use numerical simulations, which provide unique information on DOX-membrane interactions at the atomic level of resolution. In particular, we combine classical molecular dynamics simulations and free energy calculations to elucidate the mechanism of penetration of a protonated Doxorubicin molecule (DOX+) into potential liposome membranes, here modeled as lipid bilayers based on mixtures of phosphatidylcholine (PC), sphingomyelin (SM) and cholesterol lipid molecules, of different compositions and lipid phases. Moreover, we analyze DOX+ partitioning into relevant regions of SM-based lipid bilayer systems using a combination of free energy methods. Our results show that DOX+ penetration and partitioning are facilitated into less tightly packed SM-based membranes and are dependent on lipid composition. This work paves the way to further investigations of optimal formulations for lipid-based carriers, such as those associated with pH-responsive membranes.


Asunto(s)
Doxorrubicina/química , Membrana Dobles de Lípidos/química , Lípidos de la Membrana/química , Esfingomielinas/química , Colesterol/química , Doxorrubicina/farmacología , Portadores de Fármacos/química , Entropía , Humanos , Membrana Dobles de Lípidos/farmacología , Liposomas/química , Liposomas/farmacología , Lípidos de la Membrana/farmacología , Potenciales de la Membrana/efectos de los fármacos , Simulación de Dinámica Molecular , Neoplasias/tratamiento farmacológico , Esfingomielinas/farmacología
13.
Dentomaxillofac Radiol ; 51(3): 20210015, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34739351

RESUMEN

OBJECTIVES: Cleidocranial dysplasia (CCD) is a rare skeletal syndrome affecting craniofacial and dental development. As a consequence, conventional cephalometric landmarks may not be valid for CCD patients, and the appropriateness of norms used for the general population should be critically discussed. METHODS: Five patients 9- to 22-year-old (three females, two males) with CCD were included. Lateral-cephalograms, orthopantomographies, and intra-oral photos were retrospectively analysed. Lateral-cephalograms of 50 normal controls (ten for each CCD patient) matched for age and sex were selected from an online database. Cephalometric measurements of each CCD patients were compared with average values of matched controls using Wilcoxon signed-rank test for paired values (α = 0.05). RESULTS: In CCD patients, a shortening of the cranial base was present (ΔSN = -17.1 mm, p = 0.043). Thus, the mandible (ΔSNPg = +9.5°, p = 0.043) and the maxilla (ΔSNA = +11.2°, p = 0.043) showed protrusion compared to the cranial base, despite a reduced maxillary (ΔCo-A = -15.1 mm, p = 0.043) and mandibular (ΔCo-Gn = -15.2 mm, p = 0.080) length. The mandibular divergence was reduced (ΔSN/GoGn = -6.4°, p = 0.043), a reduced overbite was present (ΔOverbite = -2.9 mm, p = 0.043), and the interincisal angle was increased (ΔInterincisalAngle = +13.7°, p = 0.043), mainly due to retro-inclination of lower incisors. CONCLUSIONS: Standard cephalometric norms for the assessment of horizontal jaw position may not be applicable to CCD patients because of a reduced anterior cranial base length compared to normal subjects. Vertical relationships may not be affected, and mandibular hypodivergency was confirmed.


Asunto(s)
Displasia Cleidocraneal , Adolescente , Adulto , Cefalometría , Niño , Displasia Cleidocraneal/diagnóstico por imagen , Femenino , Humanos , Incisivo , Masculino , Mandíbula/diagnóstico por imagen , Maxilar , Estudios Retrospectivos , Adulto Joven
14.
Nanomedicine ; 7(5): 560-71, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21703989

RESUMEN

The loss of synapses and neurons in Alzheimer's disease (AD) is thought to be at least partly induced by toxic species formed by the amyloid beta (Aß) peptide; therefore, therapeutics aimed at reducing Aß toxicity could be of clinical use for treatment of AD. Liposomes are suitable vehicles for therapeutic agents and imaging probes, and a promising way of targeting the various Aß forms. We tested liposomes functionalized with phosphatidic acid, cardiolipin, or GM1 ganglioside, previously shown to have high Aß-binding capacity. Mimicking Aß-induced toxicity in mouse neuroblastoma cell lines, combined with administration of cell viability-modulating agents, we observed that functionalized liposomes rescued cell viability to different extents. We also detected rescue of the imbalance of GSK-3ß and PP2A activity, and reduction in tau phosphorylation. Thus, these liposomes appear particularly suitable for implementing further therapeutic strategies for AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Cardiolipinas/química , Gangliósido G(M1)/química , Liposomas/química , Ácidos Fosfatidicos/química , Péptidos beta-Amiloides/química , Animales , Hidrolasas de Éster Carboxílico/metabolismo , Cardiolipinas/efectos adversos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Gangliósido G(M1)/efectos adversos , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Liposomas/efectos adversos , Ratones , Ácidos Fosfatidicos/efectos adversos , Fosforilación/efectos de los fármacos , Proteínas tau/metabolismo
15.
Nanomedicine ; 7(5): 551-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21658472

RESUMEN

A promising strategy to enhance blood-brain barrier penetration by drugs is the functionalization of nanocarriers with uptake-facilitating ligands. We studied the cellular uptake, by cultured RBE4 brain capillary endothelial cells, of nanoliposomes (NLs) covalently coupled with monomer or tandem dimer of apolipoprotein E (ApoE)-derived peptides (residues 141-150), at various densities. NLs without functionalization did not show either relevant membrane accumulation or cellular uptake, as monitored by confocal microscopy and quantified by fluorescence-activated cell sorting. Functionalization with peptides mediated an efficient NLs uptake that increased with peptide density; NLs carrying monomeric peptide performed the best. Moreover, we studied the ability of ApoE-NLs to enhance the transport of a drug payload through a RBE4 cell monolayer. The permeability of a tritiated curcumin derivative was enhanced after its entrapment into ApoE-NLs, in particular those functionalized with the dimer (+83% with respect to free drug, P < 0.01). Thus, these NLs appear particularly suitable for implementing further strategies for drug brain targeting.


Asunto(s)
Apolipoproteínas E/química , Barrera Hematoencefálica/metabolismo , Sistemas de Liberación de Medicamentos , Nanopartículas/administración & dosificación , Animales , Transporte Biológico , Encéfalo/metabolismo , Línea Celular , Curcumina/farmacocinética , Células Endoteliales/metabolismo , Citometría de Flujo , Humanos , Liposomas , Microscopía Confocal , Permeabilidad , Ratas
16.
Nanomedicine ; 7(5): 541-50, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21722618

RESUMEN

The effect of various types of nanoliposomes (associated with curcumin, phosphatidic acid, cardiolipin, or GM1 ganglioside) on the aggregation of the amyloid-ß(1-42) (Aß(1-42)) peptide was investigated. Nanoliposomes incorporating curcumin (curcumin-liposomes) were prepared by adding curcumin in the lipid phase during liposome preparation, whereas curcumin surface-decorated liposomes were prepared by using a curcumin-lipid conjugate (lipid-S-curcumin liposomes) or by attaching a curcumin derivative on preformed liposomes by click chemistry (click-curcumin liposomes). The lipid ligands (phosphatidic acid, cardiolipin, or GM1) were also incorporated into nanoliposomes during their formation. All nanoliposomes with curcumin, or the curcumin derivative, were able to inhibit the formation of fibrillar and/or oligomeric Aß in vitro. Of the three forms of curcumin liposomes tested, the click-curcumin type was by far the most effective. Liposomes with lipid ligands only inhibited Aß fibril and oligomer formation at a very high ratio of liposome to peptide. Curcumin-based liposomes could be further developed as a novel treatment for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/antagonistas & inhibidores , Curcumina/administración & dosificación , Nanopartículas/administración & dosificación , Fragmentos de Péptidos/antagonistas & inhibidores , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Cardiolipinas/química , Curcumina/química , Gangliósido G(M1)/química , Humanos , Ligandos , Liposomas , Nanopartículas/química , Fragmentos de Péptidos/metabolismo , Ácidos Fosfatidicos/química
17.
Mol Neurobiol ; 58(6): 2824-2835, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33511502

RESUMEN

In light of previous results, we assessed whether liposomes functionalized with ApoE-derived peptide (mApoE) and phosphatidic acid (PA) (mApoE-PA-LIP) impacted on intracellular calcium (Ca2+) dynamics in cultured human cerebral microvascular endothelial cells (hCMEC/D3), as an in vitro human blood-brain barrier (BBB) model, and in cultured astrocytes. mApoE-PA-LIP pre-treatment actively increased both the duration and the area under the curve (A.U.C) of the ATP-evoked Ca2+ waves in cultured hCMEC/D3 cells as well as in cultured astrocytes. mApoE-PA-LIP increased the ATP-evoked intracellular Ca2+ waves even under 0 [Ca2+]e conditions, thus indicating that the increased intracellular Ca2+ response to ATP is mainly due to endogenous Ca2+ release. Indeed, when Sarco-Endoplasmic Reticulum Calcium ATPase (SERCA) activity was blocked by cyclopiazonic acid (CPA), the extracellular application of ATP failed to trigger any intracellular Ca2+ waves, indicating that metabotropic purinergic receptors (P2Y) are mainly involved in the mApoE-PA-LIP-induced increase of the Ca2+ wave triggered by ATP. In conclusion, mApoE-PA-LIP modulate intracellular Ca2+ dynamics evoked by ATP when SERCA is active through inositol-1,4,5-trisphosphate-dependent (InsP3) endoplasmic reticulum Ca2+ release. Considering that P2Y receptors represent important pharmacological targets to treat cognitive dysfunctions, and that P2Y receptors have neuroprotective effects in neuroinflammatory processes, the enhancement of purinergic signaling provided by mApoE-PA-LIP could counteract Aß-induced vasoconstriction and reduction in cerebral blood flow (CBF). Our obtained results could give an additional support to promote mApoE-PA-LIP as effective therapeutic tool for Alzheimer's disease (AD).


Asunto(s)
Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Encéfalo/patología , Señalización del Calcio , Células Endoteliales/metabolismo , Microvasos/patología , Receptores Purinérgicos/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Línea Celular , Retículo Endoplásmico/metabolismo , Células Endoteliales/efectos de los fármacos , Humanos , Indoles/farmacología , Liposomas , Ácidos Fosfatidicos/química , Ratas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
18.
Int J Pharm ; 583: 119351, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32339634

RESUMEN

Here, polyethylene glycol (PEG)-stabilized solid lipid nanoparticles (SLNs) containing Pt(IV) prodrugs derived from kiteplatin were designed and proposed as novel nanoformulations potentially useful for the treatment of glioblastoma multiforme. Four different Pt(IV) prodrugs were synthesized, starting from kiteplatin by the addition of two carboxylate ligands with different length of the alkyl chains and lipophilicity degree, and embedded in the core of PEG-stabilized SLNs composed of cetyl palmitate. The SLNs were extensively characterized by complementary optical and morphological techniques. The results proved the formation of SLNs characterized by average size under 100 nm and dependence of drug encapsulation efficiency on the lipophilicity degree of the tested Pt(IV) prodrugs. A monolayer of immortalized human cerebral microvascular endothelial cells (hCMEC/D3) was used as in vitro model of blood-brain barrier (BBB) to evaluate the ability of the SLNs to penetrate the BBB. For this purpose, optical traceable SLNs were achieved by co-incorporation of Pt(IV) prodrugs and luminescent carbon dots (C-Dots) in the SLNs. Finally, an in vitro study was performed by using a human glioblastoma cell line (U87), to investigate on the antitumor efficiency of the SLNs and on their improved ability to be cell internalized respect to the free Pt(IV) prodrugs.


Asunto(s)
Antineoplásicos/administración & dosificación , Portadores de Fármacos/administración & dosificación , Lípidos/administración & dosificación , Nanopartículas/administración & dosificación , Compuestos Organoplatinos/administración & dosificación , Polietilenglicoles/administración & dosificación , Profármacos/administración & dosificación , Antineoplásicos/química , Encéfalo/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos/química , Liberación de Fármacos , Células Endoteliales/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Humanos , Lípidos/química , Nanopartículas/química , Compuestos Organoplatinos/química , Polietilenglicoles/química , Profármacos/química , Puntos Cuánticos/administración & dosificación , Puntos Cuánticos/química
19.
Eur J Pharm Biopharm ; 149: 12-20, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32007589

RESUMEN

By functionalizing the surface of PEG-liposomes with linkers bearing quaternary ammonium compounds (QACs), we generated novel bacteria disruptors with anti-adhesive properties and reduced cytotoxicity compared to free QACs. Furthermore, QAC-functionalized liposomes are a promising platform for future drug encapsulation. The QAC (11-mercaptoundecyl)-N,N,N-trimethylammonium bromide (MTAB) was attached to maleimide-functionalized liposomes (DSPE-PEG) via thiol linker. The MTAB-functionalized liposomes were physicochemically characterized and their biological activity, in terms of anti-adherence activity and biofilm prevention in Escherichia coli were assessed. The results showed that MTAB-functionalized liposomes inhibit bacterial adherence and biofilm formation while reducing MTAB toxicity.


Asunto(s)
Antibacterianos/farmacología , Biopelículas/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Compuestos de Amonio Cuaternario/farmacología , Compuestos de Sulfhidrilo/farmacología , Antibacterianos/química , Liposomas , Maleimidas/química , Nanopartículas , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Compuestos de Amonio Cuaternario/química , Compuestos de Sulfhidrilo/química , Tensoactivos/química , Tensoactivos/farmacología
20.
Eur J Pharm Biopharm ; 142: 70-82, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31176723

RESUMEN

Nanoparticles may provide a viable way for neuroprotective drugs to cross the blood-brain barrier (BBB), which limits the passage of most drugs from the peripheral circulation to the brain. Heterotelechelic polymer prodrugs comprising a neuroprotective model drug (adenosine) and a maleimide functionality were synthesized by the "drug-initiated" approach and subsequent nitroxide exchange reaction. Nanoparticles were obtained by nanoprecipitation and exhibited high colloidal stability with diameters in the 162-185 nm range and narrow size distributions. Nanoparticles were then covalently surface-conjugated to different proteins (albumin, α2-macroglobulin and fetuin A) to test their capability of enhancing BBB translocation. Their performances in terms of endothelial permeability and cellular uptake in an in vitro BBB model were compared to that of similar nanoparticles with surface-adsorbed proteins, functionalized or not with the drug. It was shown that bare NPs (i.e., NPs not surface-functionalized with proteins) without the drug exhibited significant permeability and cellular uptake, which were further enhanced by NP surface functionalization with α2-macroglobulin. However, the presence of the drug at the polymer chain-end prevented efficient passage of all types of NPs through the BBB model, likely due to adecrease in the hydrophobicity of the nanoparticle surface and alteration of the protein binding/coupling, respectively. These results established a new and facile synthetic approach for the surface-functionalization of polymer nanoparticles for brain delivery purposes.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Nanopartículas/metabolismo , Polímeros/metabolismo , Profármacos/metabolismo , Proteínas/metabolismo , Adsorción/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Portadores de Fármacos/metabolismo , Humanos , Permeabilidad/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA