Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Int J Mol Sci ; 21(5)2020 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-32121399

RESUMEN

The binding of Aß42 peptide monomers to sphingomyelin/cholesterol (1:1 mol ratio) bilayers containing 5 mol% gangliosides (either GM1, or GT1b, or a mixture of brain gangliosides) has been assayed by density gradient ultracentrifugation. This procedure provides a direct method for measuring vesicle-bound peptides after non-bound fraction separation. This centrifugation technique has rarely been used in this context previously. The results show that gangliosides increase by about two-fold the amount of Aß42 bound to sphingomyelin/cholesterol vesicles. Complementary studies of the same systems using thioflavin T fluorescence, Langmuir monolayers or infrared spectroscopy confirm the ganglioside-dependent increased binding. Furthermore these studies reveal that gangliosides facilitate the aggregation of Aß42 giving rise to more extended ß-sheets. Thus, gangliosides have both a quantitative and a qualitative effect on the binding of Aß42 to sphingomyelin/cholesterol bilayers.


Asunto(s)
Péptidos beta-Amiloides/química , Colesterol/química , Gangliósidos/química , Fragmentos de Péptidos/química , Esfingomielinas/química , Fenómenos Biofísicos , Centrifugación por Gradiente de Densidad , Humanos , Membrana Dobles de Lípidos/química , Liposomas/química , Unión Proteica
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Biochemistry ; 47(19): 5433-40, 2008 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-18416535

RESUMEN

The influence of membrane lipid environment on the activity of GPI-anchored enzymes was investigated with human placental alkaline phosphatase reconstituted by a detergent-dialysis technique in liposomes composed of palmitoyloleoylphosphatidylcholine, alone or in mixture with lipids enriched along with the protein within lipid rafts: cholesterol, sphingomyelin, and GM1 ganglioside. The highest V max was recorded for a phosphatidylcholine/10% GM1 mixture (143 +/- 5 nmol of substrate hydrolyzed per minute per microgram of protein), while the lowest for a phosphatidylcholine/30% cholesterol mixture and for raft-mimicking 1:1:1 phosphatidylcholine/sphingolipid/cholesterol liposomes (M:M:M) (57 +/- 3 and 52 +/- 3, respectively). No significant differences in K m were detected. The protein segregation, assessed using the chemical cross-linker bis(sulfosuccinimidyl)suberate, increased with the protein:lipid ratio, within the 1:1200-1:4800 protein:lipid molar ratio range, but did not affect enzyme activity. The activity decreased when the order of the lipid bilayers was increased, higher for those containing cholesterol, as judged by steady-state fluorescence polarization of 1,6-diphenyl-1,3,5-hexatriene. Finally, the GPI-enzyme activity was affected by membrane curvature. This result was suggested by a strong inverse correlation (Pearson's correlation coefficient = 0.91; p < 0.0001) between activity and liposome diameter, measured by laser light scattering and ranging between 59 +/- 6 nm for a phosphatidylcholine/10% GM1 mixture (displaying the highest activity) and 188 +/- 25 nm for a phosphatidylcholine/30% cholesterol mixture and 185 +/- 23 nm for raft-mimicking liposomes (displaying the lowest activities). The activity-membrane curvature relationship was further confirmed by comparing the activity of proteoliposomes having different sizes but identical lipid compositions. These data open the possibility that the activity of GPI-anchored enzymes may be modulated by membrane microenvironment features, in particular by membrane curvature and cholesterol-enriched ordered microenvironments, such as those of lipid rafts.


Asunto(s)
Fosfatasa Alcalina/metabolismo , Glicosilfosfatidilinositoles/química , Glicosilfosfatidilinositoles/metabolismo , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Modelos Biológicos , Fosfatasa Alcalina/aislamiento & purificación , Anisotropía , Humanos , Cinética , Liposomas/metabolismo , Tamaño de la Partícula , Placenta/enzimología
13.
FEBS Lett ; 582(2): 215-20, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18082145

RESUMEN

Interaction of full length recombinant hamster prion protein with liposomes mimicking lipid rafts or non-raft membrane regions was studied by circular dichroism, chemical cross-linking and sucrose gradient ultracentrifugation. At pH 7.0, the protein bound palmitoyloleoylphosphatidylcholine/cholesterol/sphingomyelin/monosialoganglioside GM1 (GM1) ganglioside liposomes but not palmitoyloleoylphosphatidylcholine alone (bound/free=0.33 and 0.01, respectively), maintaining the native alpha-helical structure and monomeric form. At pH 5.0, though still binding to quaternary mixtures, in particular GM1, the protein bound also to palmitoyloleoylphosphatidylcholine (bound/free 0.35) becoming unfolded and oligomeric. The pH-dependent interaction with raft or non-raft membranes might have implication in vivo, by stabilizing or destabilizing the protein.


Asunto(s)
Concentración de Iones de Hidrógeno , Modelos Moleculares , Priones/química , Animales , Membrana Celular/metabolismo , Cricetinae , Liposomas , Conformación Proteica , Proteínas Recombinantes/química
14.
Nanomedicine (Lond) ; 13(6): 585-594, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29376461

RESUMEN

AIM: To investigate if and how the ability of liposomes, previously designed for Alzheimer's therapy, to reach the brain changes in aging/pathological conditions with respect to the healthy state. METHODS: Biodistribution and pharmacokinetics of liposomes in young or aged healthy mice and in an Alzheimer's mouse model were measured by radiochemical techniques. The expression of brain receptors and structural proteins was evaluated by Western blot. RESULTS: At equal blood levels, the amount and integrity of liposomes in the brain were dramatically lower in Alzheimer's or aged mice, with respect to young animals. These differences are likely attributable to molecular alterations in the brain vasculature. CONCLUSION: Brain alterations in pathology or aging should be considered in the design of drug delivery systems for brain targeting.


Asunto(s)
Envejecimiento/patología , Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Liposomas/administración & dosificación , Envejecimiento/efectos de los fármacos , Enfermedad de Alzheimer/patología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Humanos , Liposomas/química , Liposomas/farmacocinética , Ratones , Distribución Tisular
15.
Phytomedicine ; 42: 233-244, 2018 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-29655691

RESUMEN

BACKGROUND: Multi-target drugs have gained significant recognition for the treatment of multifactorial diseases such as depression. Under a screening study of multi-potent medicinal plants with claimed antidepressant-like activity, the phenolic-rich Annona muricata aqueous extract (AE) emerged as a moderate monoamine oxidase A (hMAO-A) inhibitor and a strong hydrogen peroxide (H2O2) scavenger. PURPOSE: In order to protect this extract from gastrointestinal biotransformation and to improve its permeability across the blood-brain barrier (BBB), four phospholipid nanoformulations of liposomes and phytosomes functionalized with a peptide ligand promoting BBB crossing were produced. METHODS: AE and nanoformulations were characterized by HPLC-DAD-ESI-MSn, HPLC-DAD, spectrophotometric, fluorescence and dynamic light scattering methods. Cytotoxicity and permeability studies were carried out using an in vitro transwell model of the BBB, composed of immortalized human microvascular endothelial cells (hCMEC/D3), and in vitro hMAO-A inhibition and H2O2 scavenging activities were performed with all samples. RESULTS: The encapsulation/binding of AE was more efficient with phytosomes, while liposomes were more stable, displaying a slower extract release over time. In general, phytosomes were less toxic than liposomes in hCMEC/D3 cells and, when present, cholesterol improved the permeability across the cell monolayer of all tested nanoformulations. All nanoformulations conserved the antioxidant potential of AE, while phosphatidylcholine interfered with MAO-A inhibition assay. CONCLUSIONS: Overall, phytosome formulations registered the best performance in terms of binding efficiency, enzyme inhibition and scavenging activity, thus representing a promising multipotent phenolic-rich nanoshuttle for future in vivo depression treatment.


Asunto(s)
Annona/química , Antioxidantes/farmacología , Portadores de Fármacos/química , Liposomas/química , Extractos Vegetales/administración & dosificación , Barrera Hematoencefálica/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Portadores de Fármacos/administración & dosificación , Células Endoteliales/efectos de los fármacos , Depuradores de Radicales Libres/farmacología , Humanos , Peróxido de Hidrógeno/metabolismo , Liposomas/administración & dosificación , Inhibidores de la Monoaminooxidasa/farmacología , Nanoestructuras/administración & dosificación , Nanoestructuras/química , Fenoles/farmacología , Extractos Vegetales/farmacología , Espectrometría de Masa por Ionización de Electrospray
16.
Int J Nanomedicine ; 13: 4059-4071, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30034232

RESUMEN

PURPOSE: Nanotechnologies turned out to be promising in the development of diagnostic and therapeutic approaches toward neurodegenerative disorders. However, only a very scant number of nanodevices until now proved to be effective on preclinical animal models. Although specific tests in vivo are available to assess the potential toxicity of these nanodevices on cognitive functions, those to evaluate their biosafety in vitro on neurons are still to be improved. MATERIALS AND METHODS: We utilized the patch-clamp technique on primary cultures of cortical neural cells isolated from neonatal rats, aiming to evaluate their electrical properties after the incubation with liposomes (mApoE-PA-LIPs), previously proved able to cross the blood-brain barrier and to be effective on mouse models of Alzheimer's disease (AD), both in the absence and in the presence of ß-amyloid peptide oligomers. RESULTS: Data show a high degree of biocompatibility, evaluated by lactate dehydrogenase (LDH) release and MTT assay, and the lack of cellular internalization. After the incubation with mApoE-PA-LIPs, neuronal membranes show an increase in the input resistance (from 724.14±76 MΩ in untreated population to 886.06±86 MΩ in the treated one), a reduction in the rheobase current (from 29.6±3 to 24.2±3 pA in untreated and treated, respectively), and an increase of the firing frequency, consistent with an ultimate increase in intrinsic excitability. Data obtained after co-incubation of mApoE-PA-LIPs with ß-amyloid peptide oligomers suggest a retention of liposome efficacy. CONCLUSION: These data suggest the ability of liposomes to modulate neuronal electrical properties and are compatible with the previously demonstrated amelioration of cognitive functions induced by treatment of AD mice with liposomes. We conclude that this electrophysiological approach could represent a useful tool for nanomedicine to evaluate the effect of nanoparticles on intrinsic neuronal excitability.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Neuronas/metabolismo , Potenciales de Acción , Péptidos beta-Amiloides/metabolismo , Animales , Animales Recién Nacidos , Apolipoproteínas E/metabolismo , Materiales Biocompatibles/química , Supervivencia Celular , Células Cultivadas , Endocitosis , Liposomas , Masculino , Ratones , Nanopartículas/química , Ácidos Fosfatidicos/química , Ratas
17.
J Control Release ; 258: 121-129, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28501671

RESUMEN

The failure of clinical trials largely focused on mild to moderate stages of Alzheimer disease has suggested to the scientific community that the effectiveness of Amyloid-ß (Aß)-centered treatments should be evaluated starting as early as possible, well before irreversible brain damage has occurred. Accordingly, also the preclinical development of new therapies should be carried out taking into account this suggestion. In the present investigation we evaluated the efficacy of a treatment with liposomes multifunctionalized for crossing the blood-brain barrier and targeting Aß, carried out on young APP/PS1 Tg mice, taken as a model of pre-symptomatic disease stage. Liposomes were administered once a week to Tg mice for 7months, starting at the age of 5months and up to the age of 12 when they display AD-like cognitive and brain biochemical/anatomical features. The treatment prevented the onset of the long-term memory impairment and slowed down the deposition of brain Aß; at anatomical level, prevented both ventricle enlargement and entorhinal cortex thickness reduction, otherwise occurring in untreated mice. Strikingly, these effects were maintained 3months after treatment discontinuation. An increase of Aß levels in the liver was detected at the end of the treatment, then followed also by reduction of brain Amyloid Precursor Protein and increase of Aß-degrading enzymes. These results suggest that the treatment promotes brain Aß clearance by a peripheral 'sink' effect and ultimately affects Aß turnover in the brain. Worth of note, the treatment was apparently not toxic for all the organs analyzed, in particular for brain, as suggested by the lower brain TNF-α and MDA levels, and by higher level of SOD activity in treated mice. Together, these findings promote a very early treatment with multi-functional liposomes as a well-tolerated nanomedicine-based approach, potentially suitable for a disease-modifying therapy of AD, able to delay or prevent relevant features of the disease.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Apolipoproteínas E/uso terapéutico , Encéfalo/efectos de los fármacos , Liposomas/uso terapéutico , Trastornos de la Memoria/prevención & control , Ácidos Fosfatidicos/uso terapéutico , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Apolipoproteínas E/administración & dosificación , Apolipoproteínas E/química , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Sistemas de Liberación de Medicamentos , Liposomas/administración & dosificación , Liposomas/química , Masculino , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Ratones Transgénicos , Ácidos Fosfatidicos/administración & dosificación , Ácidos Fosfatidicos/química
18.
Neurochem Int ; 108: 60-65, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28238790

RESUMEN

The accumulation of extracellular amyloid beta (Abeta42) both in brain and in cerebral vessels characterizes Alzheimer's disease (AD) pathogenesis. Recently, the possibility to functionalize nanoparticles (NPs) surface with Abeta42 binding molecules, making them suitable tools for reducing Abeta42 burden has been shown effective in models of AD. Aim of this work consisted in proving that NPs might be effective in sequestering Abeta42 in biological fluids, such as CSF and plasma. This demonstration is extremely important considering that these Abeta42 pools are in continuum with the brain parenchyma with drainage of Abeta from interstitial brain tissue to blood vessel and plasma. In this work, liposomes (LIP) were functionalized as previously shown in order to promote high-affinity Abeta binding, i.e., either with, phosphatidic acid (PA), or a modified Apolipoprotein E-derived peptide (mApo), or with a curcumin derivative (TREG); Abeta42 levels were determined by ELISA in CSF and plasma samples. mApo-PA-LIP (25 and 250 µM) mildly albeit significantly sequestered Abeta42 proteins in CSF samples obtained from healthy subjects (p < 0.01). Analogously a significant binding (∼20%) of Abeta42 (p < 0.001) was demonstrated following exposure to all functionalized liposomes in plasma samples obtained from selected AD or Down's syndrome patients expressing high levels of Abeta42. The same results were obtained by quantifying Abeta42 content after removal of liposome-bound Abeta by using gel filtration chromatography or ultracentrifugation on a discontinuous sucrose density gradient. In conclusion, we demonstrate that functionalized liposomes significantly sequester Abeta42 in human biological fluids. These data may be critical for future in vivo administration tests using NPs for promoting sink effect.


Asunto(s)
Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/líquido cefalorraquídeo , Péptidos beta-Amiloides/sangre , Péptidos beta-Amiloides/líquido cefalorraquídeo , Liposomas/metabolismo , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/líquido cefalorraquídeo , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Femenino , Humanos , Masculino
19.
Neurobiol Aging ; 57: 84-94, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28605642

RESUMEN

In this study, we evaluated the anti-amyloid effect of functionalized nanoliposomes (mApoE-PA-LIP) in a mouse model of Alzheimer's disease with use of positron emission tomography and ß-amyloid (Aß)-targeted tracer [11C]Pittsburgh compound B ([11C]PIB). APP23 mice were injected with mApoE-PA-LIP or saline (3 times per week for 3 weeks) and [11C]PIB imaging was performed at baseline, after the treatment and after 3 months follow-up period, accompanied by Aß immunohistochemistry and ELISA. After the treatment, [11C]PIB binding ratios between mApoE-PA-LIP and saline groups were equivalent in all analyzed brain regions; however, in the saline group, binding ratios increased from the baseline, whereas no increase was detected in the mApoE-PA-LIP group. During the additional follow-up, [11C]PIB binding increased significantly from baseline in both groups, and binding ratios correlated with the immunohistochemically defined Aß load. This study further supports the use of [11C]PIB positron emission tomography imaging as a biomarker of Aß deposition in APP23 mice and highlights the benefits of noninvasive follow-up, that is, using baseline data for animal stratification and normalization of treatment effects to baseline values, for future anti-amyloid treatment studies.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/metabolismo , Compuestos de Anilina , Liposomas/administración & dosificación , Liposomas/uso terapéutico , Tomografía de Emisión de Positrones , Tiazoles , Enfermedad de Alzheimer/metabolismo , Animales , Radioisótopos de Carbono , Modelos Animales de Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Liposomas/farmacología , Masculino , Ratones Transgénicos , Nanopartículas , Radiofármacos
20.
J Pharm Sci ; 105(1): 276-83, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26852859

RESUMEN

Surface functionalization with antitransferrin receptor (TfR) mAbs has been suggested as the strategy to enhance the transfer of nanoparticles (NPs) across the blood-brain barrier (BBB) and to carry nonpermeant drugs from the blood into the brain. However, the efficiency of BBB crossing is currently too poor to be used in vivo. In the present investigation, we compared 6 different murine mAbs specific for different epitopes of the human TfR to identify the best performing one for the functionalization of NPs. For this purpose, we compared the ability of mAbs to cross an in vitro BBB model made of human brain capillary endothelial cells (hCMEC/D3). Liposomes functionalized with the best performing mAb (MYBE/4C1) were uptaken, crossed the BBB in vitro, and facilitated the BBB in vitro passage of doxorubicin, an anticancer drug, 3.9 folds more than liposomes functionalized with a nonspecific IgG, as assessed by confocal microscopy, radiochemical techniques, and fluorescence, and did not modify the cell monolayer structural or functional properties. These results show that MYBE/4C1 antihuman TfR mAb is a powerful resource for the enhancement of BBB crossing of NPs and is therefore potentially useful in the treatment of neurologic diseases and disorders including brain carcinomas.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Receptores de Transferrina/antagonistas & inhibidores , Animales , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/farmacocinética , Capilares/efectos de los fármacos , Capilares/metabolismo , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Sistemas de Liberación de Medicamentos , Células Endoteliales , Epítopos , Humanos , Inmunoglobulina G/química , Liposomas , Ratones , Tamaño de la Partícula
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA