Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Prog Brain Res ; 245: 57-88, 2019.
Article in English | MEDLINE | ID: mdl-30961872

ABSTRACT

The treatment of Alzheimer's disease (AD) is up to today one of the most unsuccessful examples of biomedical science. Despite the high number of literature evidences detailing the multifactorial and complex etiopathology of AD, no cure is yet present on the market and the available treatments are only symptomatic. The reasons could be ascribed on two main factors: (i) lack of ability of the majority of drugs to cross the blood-brain barrier (BBB), thus excluding the brain for any successful therapy; (ii) lack of selectivity and specificity of drugs, decreasing the efficacy of even potent anti-AD drugs. The exploitation of specifically engineered nanomedicines planned to cross the BBB and to target the most "hot" site of action (i.e., ß-amyloid) is one of the most interesting innovations in drug delivery and could reasonably represent an promising choice for possible treatments and even early-diagnosis of AD. In this chapter, we therefore outline the most talented approaches in AD treatment with a specific focus on the main advantages/drawbacks and future possible translation to clinic application.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/drug effects , Blood-Brain Barrier , Nanomedicine/methods , Neuroprotective Agents/administration & dosage , Nootropic Agents/administration & dosage , Animals , Humans
2.
Int J Pharm ; 543(1-2): 300-310, 2018 May 30.
Article in English | MEDLINE | ID: mdl-29608954

ABSTRACT

Restoration of the Chol homeostasis in the Central Nervous System (CNS) could be beneficial for the treatment of Huntington's Disease (HD), a progressive, fatal, adult-onset, neurodegenerative disorder. Unfortunately, Chol is unable to cross the blood-brain barrier (BBB), thus a novel strategy for a targeted delivery of Chol into the brain is highly desired. This article aims to investigate the production of hybrid nanoparticles composed by Chol and PLGA (MIX-NPs) modified with g7 ligand for BBB crossing. We described the impact of ratio between components (Chol and PLGA) and formulation process (nanoprecipitation or single emulsion process) on physico-chemical and structural characteristics, we tested MIX-NPs in vitro using primary hippocampal cell cultures evaluating possible toxicity, uptake, and the ability to influence excitatory synaptic receptors. Our results elucidated that both formulation processes produce MIX-NPs with a Chol content higher that 40%, meaning that Chol is a structural particle component and active compound at the same time. The formulation strategy impacted the architecture and reorganization of components leading to some differences in Chol availability between the two types of g7 MIX-NPs. Our results identified that both kinds of MIX-NPs are efficiently taken up by neurons, able to escape lysosomes and release Chol into the cells resulting in an efficient modification in expression of synaptic receptors that could be beneficial in HD.


Subject(s)
Cholesterol/chemistry , Drug Delivery Systems , Glycopeptides/chemistry , Lactic Acid/chemistry , Nanoparticles/chemistry , Polyglycolic Acid/chemistry , Animals , Cell Survival/drug effects , Cells, Cultured , Cholesterol/administration & dosage , Embryo, Mammalian , Glycopeptides/administration & dosage , Hippocampus/cytology , Lactic Acid/administration & dosage , Nanoparticles/administration & dosage , Neurons/drug effects , Neurons/metabolism , Poloxamer/chemistry , Polyglycolic Acid/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer , Polyvinyl Alcohol/chemistry , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Surface-Active Agents/administration & dosage , Surface-Active Agents/chemistry
3.
J Trace Elem Med Biol ; 49: 210-221, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29325805

ABSTRACT

A local dyshomeostasis of zinc ions in the vicinity of amyloid aggregates has been proposed in Alzheimer's disease (AD) due to the sequestration of zinc in senile plaques. While an increase in zinc levels may promote the aggregation of amyloid beta (Aß), increased brain zinc might also be beneficial rescuing some pathological alterations caused by local zinc deficiency. For example, increased Aß degradation by metalloproteinases, and a reduction in inflammation can be hypothesized. In addition, zinc may allow a stabilization of the number of synapses in AD brains. Thus, to evaluate whether altering zinc-levels within the brain is a promising new target for the prevention and treatment of AD, we employed novel zinc loaded nanoparticles able to deliver zinc into the brain across the blood-brain barrier. We performed in vivo studies using wild type (WT) and APP23 mice to assess plaque load, inflammatory status and synapse loss. Furthermore, we performed behavioral analyses. After chronically injecting these nanoparticles for 14 days, our results show a significant reduction in plaque size and effects on the pro-inflammatory cytokines IL-6 and IL-18. On behavioral level we could not detect negative effects of increased brain zinc levels in APP23 mice and treatment with g7-NP-Zn normalized the observed hyperlocomotion of APP23 mice. Therefore, we conclude that a targeted increase in brain zinc levels may have beneficial effects in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Nanoparticles/chemistry , Polymers/chemistry , Zinc/metabolism , Zinc/therapeutic use , Alzheimer Disease/drug therapy , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Disease Models, Animal , Female , Male , Mice , Nanoparticles/therapeutic use , Real-Time Polymerase Chain Reaction , Zinc/administration & dosage
4.
Nanomedicine (Lond) ; 13(4): 407-422, 2018 02.
Article in English | MEDLINE | ID: mdl-29345202

ABSTRACT

AIM: The investigation on protein coronas (PCs) adsorbed onto nanoparticle (NP) surface is representing an open issue due to difficulties in detection and clear isolation of the adsorbed proteins. In this study, we investigated protocols able to isolate the compositions of PCs of three polymeric NPs. MATERIALS & METHODS: Unfunctionalized NPs and two functionalized NPs were considered as proof-of-concept for the qualitative and semiquantitative analysis of both the corona levels (stably or weakly adsorbed coronas [SC/WC]) of these different nanocarriers. RESULTS: The protocols applied were able to discriminate between the SC and WC. In particular, experimental results indicated that stably adsorbed coronas are prevalently composed by ApoE, while WC by albumin in all the NPs. Otherwise, some differences in WC could be correlated with surface functionalization. CONCLUSION: This experimental approach allows characterizing the whole PCs, proposing a protocol for isolation of different types of proteins composing PCs.


Subject(s)
Antibodies, Monoclonal/chemistry , Nanoparticles/chemistry , Oligopeptides/chemistry , Protein Corona/analysis , Adsorption , Albumins/chemistry , Apolipoproteins E/chemistry , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Proof of Concept Study , Surface Properties
5.
Int Rev Neurobiol ; 137: 1-28, 2017.
Article in English | MEDLINE | ID: mdl-29132540

ABSTRACT

Brain diseases and injuries are growing to be one of the most deadly and costly medical conditions in the world. Unfortunately, current treatments are incapable of ameliorating the symptoms let alone curing the diseases. Many brain diseases have been linked to a loss of function in a protein or enzyme, increasing research for improving their delivery. This is no easy task due to the delicate nature of proteins and enzymes in biological conditions, as well as the many barriers that exist in the body ranging from those in circulation to the more specific barriers to enter the brain. Several main techniques are being used (physical delivery, protein/enzyme conjugates, and nanoparticle delivery) to overcome these barriers and create new therapeutics. This review will cover recently published data and highlights the benefits and deficits of possible new protein or enzyme therapeutics for brain diseases.


Subject(s)
Brain Diseases/therapy , Drug Delivery Systems/methods , Proteins/administration & dosage , Humans , Nanoparticles/administration & dosage
6.
Int J Pharm ; 526(1-2): 413-424, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28495580

ABSTRACT

The formation of extracellular aggregates built up by deposits of ß-amyloid (Aß) is a hallmark of Alzheimer's disease (AD). Curcumin has been reported to display anti-amyloidogenic activity, not only by inhibiting the formation of new Aß aggregates, but also by disaggregating existing ones. However, the uptake of Curcumin into the brain is severely restricted by its low ability to cross the blood-brain barrier (BBB). Therefore, novel strategies for a targeted delivery of Curcumin into the brain are highly desired. Here, we encapsulated Curcumin as active ingredient in PLGA (polylactide-co-glycolic-acid) nanoparticles (NPs), modified with g7 ligand for BBB crossing. We performed in depth analyses of possible toxicity of these NPs, uptake, and, foremost, their ability to influence Aß pathology in vitro using primary hippocampal cell cultures. Our results show no apparent toxicity of the formulated NPs, but a significant decrease of Aß aggregates in response to Curcumin loaded NPs. We thus conclude that brain delivery of Curcumin using BBB crossing NPs is a promising future approach in the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/drug effects , Blood-Brain Barrier , Curcumin/administration & dosage , Nanoparticles/chemistry , Biological Transport , Humans
7.
Article in English | MEDLINE | ID: mdl-28296346

ABSTRACT

Nanoparticles (NPs) represent one of the most promising tools for drug-targeting and drug-delivery. However, a deeper understanding of the complex dynamics that happen after their in vivo administration is required. Particularly, plasma proteins tend to associate to NPs, forming a new surface named the 'protein corona' (PC). This surface is the most exposed as the 'visible side' of NPs and therefore, can have a strong impact on NP biodistribution, targeting efficacy and also toxicity. The PC consists of two poorly delimited layers, known as 'hard corona' (HC) and 'soft corona' (SC), that are affected by the complexity of the environment and the formed protein-surface equilibrium during in vivo blood circulation. The HC corona is formed by proteins strongly associated to the NPs, while the SC is an outer layer consisting of loosely bound proteins. Several studies attempted to investigate the HC, which is easier to be isolated, but yielded poor reproducibility, due to varying experimental conditions. As a consequence, full mapping of the HC for different NPs is still lacking. Moreover, the current knowledge on the SC, which may play a major role in the 'first' interaction of NPs once in vivo, is very limited, mainly due to the difficulties in preserving it after purification. Therefore, multi-disciplinary approaches leading to the obtainment of a major number of information about the PC and its properties is strongly needed to fully understand its impact and to better support a more safety and conscious application of nanotechnology in medicine. WIREs Nanomed Nanobiotechnol 2017, 9:e1467. doi: 10.1002/wnan.1467 For further resources related to this article, please visit the WIREs website.


Subject(s)
Nanoparticles/chemistry , Protein Corona/chemistry , Hardness , Humans , Nanotechnology , Protein Conformation
8.
Expert Opin Drug Deliv ; 14(7): 825-840, 2017 07.
Article in English | MEDLINE | ID: mdl-27690258

ABSTRACT

INTRODUCTION: New frontiers in nanomedicine are moving towards the research of new biomaterials. Apoferritin (APO), is a uniform regular self-assemblies nano-sized protein with excellent biocompatibility and a unique structure that affords it the ability to stabilize small active molecules in its inner core. Areas covered: APO can be loaded by applying a passive process (mainly used for ions and metals) or by a unique formulative approach based on disassemby/reassembly process. In this article, we aim to organize the experimental evidence provided by a number of studies on the loading, release and targeting. Attention is initially focused on the most investigated antineoplastic drug and contrast agents up to the most recent application in gene therapy. Expert opinion: Various preclinical studies have demonstrated that APO improved the potency and selectivity of some chemotherapeutics. However, in order to translate the use of APO into therapy, some issues must be solved, especially regarding the reproducibility of the loading protocol used, the optimization of nanocarrier characterization, detailed understanding of the final structure of loaded APO, and the real mechanism and timing of drug release.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Delivery Systems , Ferritins/administration & dosage , Nanostructures/administration & dosage , Animals , Antineoplastic Agents/chemistry , Drug Liberation , Ferritins/chemistry , Ferritins/pharmacokinetics , Humans , Nanomedicine , Nanostructures/chemistry , Neoplasms/metabolism
9.
CNS Neurol Disord Drug Targets ; 15(9): 1079-1091, 2016.
Article in English | MEDLINE | ID: mdl-27633786

ABSTRACT

The research of efficacious non-invasive therapies for the treatment of brain diseases represents a huge challenge, as people affected by disorders of the central nervous system (CNS) will significantly increase. Moreover, the blood-brain barrier is a key factor in hampering a number of effective drugs to reach the CNS. This review is therefore focusing on possible interventions of nanomedicine-based approaches in selected diseases affecting the CNS. A wide overview of the most outstanding results on preclinical evaluations of the potential of nanomedicine in brain diseases (i.e. brain tumor, Alzheimer, Parkinson, epilepsy and others) is given, with highlights on the data with relevant interest and real possibility in translation from bench-to-bedside. Moreover, a critical evaluation on the rationale in planning nanosystems to target specific brain pathologies is described, opening the path to a more structured and pathology-tailored design of nanocarriers.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Central Nervous System Agents/administration & dosage , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/metabolism , Drug Delivery Systems , Animals , Humans , Nanomedicine/methods
11.
Tissue Barriers ; 4(1): e1153568, 2016.
Article in English | MEDLINE | ID: mdl-27141426

ABSTRACT

While the role of the blood-brain barrier (BBB) is increasingly recognized in the (development of treatments targeting neurodegenerative disorders, to date, few strategies exist that enable drug delivery of non-BBB crossing molecules directly to their site of action, the brain. However, the recent advent of Nanomedicines may provide a potent tool to implement CNS targeted delivery of active compounds. Approaches for BBB crossing are deeply investigated in relation to the pathology: among the main important diseases of the CNS, this review focuses on the application of nanomedicines to neurodegenerative disorders (Alzheimer, Parkinson and Huntington's Disease) and to other brain pathologies as epilepsy, infectious diseases, multiple sclerosis, lysosomal storage disorders, strokes.


Subject(s)
Blood-Brain Barrier/metabolism , Nanoparticles/metabolism , Animals , Blood-Brain Barrier/drug effects , Capillary Permeability , Central Nervous System Diseases/drug therapy , Drug Carriers/pharmacokinetics , Humans
12.
PLoS One ; 11(5): e0156452, 2016.
Article in English | MEDLINE | ID: mdl-27228099

ABSTRACT

Lysosomal Storage Disorders (LSDs) are a group of metabolic syndromes, each one due to the deficit of one lysosomal enzyme. Many LSDs affect most of the organ systems and overall about 75% of the patients present neurological impairment. Enzyme Replacement Therapy, although determining some systemic clinical improvements, is ineffective on the CNS disease, due to enzymes' inability to cross the blood-brain barrier (BBB). With the aim to deliver the therapeutic enzymes across the BBB, we here assayed biodegradable and biocompatible PLGA-nanoparticles (NPs) in two murine models for LSDs, Mucopolysaccharidosis type I and II (MPS I and MPS II). PLGA-NPs were modified with a 7-aminoacid glycopeptide (g7), yet demonstrated to be able to deliver low molecular weight (MW) molecules across the BBB in rodents. We specifically investigated, for the first time, the g7-NPs ability to transfer a model drug (FITC-albumin) with a high MW, comparable to the enzymes to be delivered for LSDs brain therapy. In vivo experiments, conducted on wild-type mice and knockout mouse models for MPS I and II, also included a whole series of control injections to obtain a broad preliminary view of the procedure efficiency. Results clearly showed efficient BBB crossing of albumin in all injected mice, underlying the ability of NPs to deliver high MW molecules to the brain. These results encourage successful experiments with enzyme-loaded g7-NPs to deliver sufficient amounts of the drug to the brain district on LSDs, where exerting a corrective effect on the pathological phenotype.


Subject(s)
Blood-Brain Barrier/metabolism , Drug Carriers , Enzyme Replacement Therapy/methods , Mucopolysaccharidosis II/drug therapy , Mucopolysaccharidosis I/drug therapy , Nanoparticles , Albumins/chemistry , Albumins/pharmacokinetics , Albumins/pharmacology , Animals , Disease Models, Animal , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Fluorescein-5-isothiocyanate/chemistry , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescein-5-isothiocyanate/pharmacology , Lactic Acid/chemistry , Lactic Acid/pharmacokinetics , Lactic Acid/pharmacology , Mice , Mice, Knockout , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/pathology , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/metabolism , Mucopolysaccharidosis II/pathology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacokinetics , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer
13.
Eur J Pharm Biopharm ; 99: 7-17, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26625717

ABSTRACT

Silencing of the B lymphocyte-induced maturation protein 1 (Blimp-1), a pivotal transcriptional regulator during terminal differentiation of B cells into plasma cells with siRNAs is under investigation as novel therapeutic approach in Primary Effusion Lymphoma (PEL), a HHV-8 related and aggressive B cell Lymphoma currently lacking of an efficacious therapeutic approach. The clinical application of small interfering RNA (siRNA) in cancer therapy is limited by the lack of an efficient systemic siRNA delivery system. In this study we aim to develop pegylated siRNA lipoplexes formed using the cationic lipid DOTAP and DSPE-PEG2000, capable to effectively stabilize anti-Blimp-1 siRNA and suitable for systemic administration. Two types of pegylated lipoplexes using a classic (C-PEG Lipoplexes) or a post-pegylation method (P-PEG-Lipoplexes) were formulated and compared in their physicochemical properties (size, zeta potential, morphology and structure) and efficiency on PEL cell lines. A stable siRNAs protection was obtained with post pegylation approach (2% molar of DSPE-PEG2000 with respect to lipid) resulting in structures with diameters of 300 nm and a complexation efficiency higher that 80% (0.08 nmol/10 nmol of lipid). In vitro studies on PEL cell lines suggested that empty liposomes were characterized by a low cell toxicity also after PEG modification (cell viability and cell density over 85% after treatment with 10 µM of lipid). We demonstrated that P-PEG-Lipoplexes were able to significantly reduce the levels of BLIMP-1 protein leading to reduction of viability (less that 15% after transfection with 100 nM of complexed siRNAs) and activation of apoptosis. In vitro efficiency encourages us to further test the in vivo potential of P-PEG-Lipoplexes in PEL therapy.


Subject(s)
Gene Silencing , Genetic Therapy/methods , Lymphoma, Primary Effusion/genetics , Polyethylene Glycols/administration & dosage , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Humans , Liposomes , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/therapy , Positive Regulatory Domain I-Binding Factor 1 , RNA, Small Interfering/administration & dosage , Repressor Proteins/antagonists & inhibitors
14.
EMBO Mol Med ; 7(12): 1547-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26589247

ABSTRACT

Brain cholesterol biosynthesis and cholesterol levels are reduced in mouse models of Huntington's disease (HD), suggesting that locally synthesized, newly formed cholesterol is less available to neurons. This may be detrimental for neuronal function, especially given that locally synthesized cholesterol is implicated in synapse integrity and remodeling. Here, we used biodegradable and biocompatible polymeric nanoparticles (NPs) modified with glycopeptides (g7) and loaded with cholesterol (g7-NPs-Chol), which per se is not blood-brain barrier (BBB) permeable, to obtain high-rate cholesterol delivery into the brain after intraperitoneal injection in HD mice. We report that g7-NPs, in contrast to unmodified NPs, efficiently crossed the BBB and localized in glial and neuronal cells in different brain regions. We also found that repeated systemic delivery of g7-NPs-Chol rescued synaptic and cognitive dysfunction and partially improved global activity in HD mice. These results demonstrate that cholesterol supplementation to the HD brain reverses functional alterations associated with HD and highlight the potential of this new drug-administration route to the diseased brain.


Subject(s)
Cholesterol/therapeutic use , Cognition/drug effects , Huntington Disease , Nanoparticles , Neurons/physiology , Synapses/physiology , Animals , Blood-Brain Barrier , Disease Models, Animal , Huntington Disease/physiopathology , Huntington Disease/therapy , Mice , Neurons/drug effects , Synapses/drug effects
15.
CNS Neurol Disord Drug Targets ; 14(8): 1041-53, 2015.
Article in English | MEDLINE | ID: mdl-26295815

ABSTRACT

A dyshomeostasis of zinc ions has been reported for many psychiatric and neurodegenerative disorders including schizophrenia, attention deficit hyperactivity disorder, depression, autism, Parkinson's and Alzheimer's disease. Furthermore, alterations in zinc-levels have been associated with seizures and traumatic brain injury. Thus, altering zinclevels within the brain is emerging as a new target for the prevention and treatment of psychiatric and neurological diseases. However, given the restriction of zinc uptake into the brain by the blood-brain barrier, methods for controlled regulation and manipulation of zinc concentrations within the brain are rare. Here, we performed in vivo studies investigating the possibility of brain targeted zinc delivery using zinc-loaded nanoparticles which are able to cross the blood-brain barrier. After injecting these nanoparticles, we analyzed the regional and time-dependent distribution of zinc and nanoparticles within the brain. Moreover, we evaluated whether the presence of zinc-loaded nanoparticles alters the expression of zinc sensitive genes and proteins such as metallothioneins and zinc transporters and quantified possible toxic effects. Our results show that zinc loaded g7 nanoparticles offer a promising approach as a novel non - invasive method to selectively enrich zinc in the brain within a small amount of time.


Subject(s)
Brain/drug effects , Central Nervous System Agents/administration & dosage , Drug Carriers , Nanoparticles , Zinc/administration & dosage , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Cations, Divalent/administration & dosage , Cations, Divalent/pharmacokinetics , Cations, Divalent/toxicity , Central Nervous System Agents/pharmacokinetics , Central Nervous System Agents/toxicity , Drug Carriers/chemistry , Drug Carriers/toxicity , Drug Evaluation, Preclinical , Glycopeptides/chemistry , Glycopeptides/toxicity , Immunohistochemistry , Lactic Acid/chemistry , Lactic Acid/toxicity , Mice, Inbred BALB C , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/metabolism , Nanoparticles/chemistry , Nanoparticles/toxicity , Polyglycolic Acid/chemistry , Polyglycolic Acid/toxicity , Polylactic Acid-Polyglycolic Acid Copolymer , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Zinc/pharmacokinetics , Zinc/toxicity
17.
Pharmaceutics ; 7(2): 74-89, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26102358

ABSTRACT

The success of nanomedicine as a new strategy for drug delivery and targeting prompted the interest in developing approaches toward basic and clinical neuroscience. Despite enormous advances on brain research, central nervous system (CNS) disorders remain the world's leading cause of disability, in part due to the inability of the majority of drugs to reach the brain parenchyma. Many attempts to use nanomedicines as CNS drug delivery systems (DDS) were made; among the various non-invasive approaches, nanoparticulate carriers and, particularly, polymeric nanoparticles (NPs) seem to be the most interesting strategies. In particular, the ability of poly-lactide-co-glycolide NPs (PLGA-NPs) specifically engineered with a glycopeptide (g7), conferring to NPs' ability to cross the blood brain barrier (BBB) in rodents at a concentration of up to 10% of the injected dose, was demonstrated in previous studies using different routes of administrations. Most of the evidence on NP uptake mechanisms reported in the literature about intracellular pathways and processes of cell entry is based on in vitro studies. Therefore, beside the particular attention devoted to increasing the knowledge of the rate of in vivo BBB crossing of nanocarriers, the subsequent exocytosis in the brain compartments, their fate and trafficking in the brain surely represent major topics in this field.

18.
Mol Neurobiol ; 52(2): 899-912, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26108180

ABSTRACT

Cerebrolysin is a peptide mixture able to ameliorate symptomatology and delay progression of neurological disorders such as Alzheimer's disease and dementia. The administration of this drug in humans presents several criticisms due to its short half-life, poor stability, and high doses needed to achieve the effect. This paper investigates the potential of polylactic-co-glycolide (PLGA) nanoparticles (NPs) as sustained release systems for iv administration of cerebrolysin in normal and brain injured rats. NPs were prepared by water-in-oil-in-water (w/o/w) double emulsion technique and characterized by light scattering for mean size and zeta potential and by scanning electron microscopy (SEM) for surface morphology. The NPs produced by double sonication under cooling at 60 W for 45 s, 12 mL of 1 % w:v of PVA, and 1:0.6 w:w drug/PLGA ratio (C-NPs4) displayed an adequate loading of drug (24 ± 1 mg/100 mg of NPs), zeta potential value (-13 mV), and average diameters (ranged from 250 to 330 nm) suitable to iv administration. SEM images suggested that cerebrolysin was molecularly dispersed into matricial systems and partially adhered to the NP surface. A biphasic release with an initial burst effect followed by sustained release over 24 h was observed. Long-term stability both at room and at low temperature of freeze-dried NPs was investigated. To gain deeper insight into NP stability after in vivo administration, the stability of the best NP formulation was also tested in serum. These PLGA NPs loaded with cerebrolysin were able to reduce brain pathology following traumatic brain injury. However, the size, the polydispersivity, and the surface properties of sample were significantly affected by the incubation time and the serum concentration.


Subject(s)
Amino Acids/administration & dosage , Brain Injuries/drug therapy , Lactic Acid , Nanoparticles , Neuroprotective Agents/administration & dosage , Polyglycolic Acid , Amino Acids/blood , Amino Acids/chemistry , Amino Acids/therapeutic use , Animals , Blood-Brain Barrier , Brain Edema/etiology , Brain Edema/physiopathology , Brain Injuries/complications , Brain Injuries/physiopathology , Delayed-Action Preparations , Drug Evaluation, Preclinical , Drug Stability , Drug Storage , Emulsions , Freeze Drying , Hydrophobic and Hydrophilic Interactions , Male , Microscopy, Electron, Scanning , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Neuroprotective Agents/blood , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Osmolar Concentration , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Time Factors , Trehalose/chemistry
19.
Nanomedicine (Lond) ; 10(11): 1735-50, 2015.
Article in English | MEDLINE | ID: mdl-26080696

ABSTRACT

AIM: Drug targeting to the CNS is challenging due to the presence of blood-brain barrier. We investigated chitosan (Cs) nanoparticles (NPs) as drug transporter system across the blood-brain barrier, based on mAb OX26 modified Cs. MATERIALS & METHODS: Cs NPs functionalized with PEG, modified and unmodified with OX26 (Cs-PEG-OX26) were prepared and chemico-physically characterized. These NPs were administered (intraperitoneal) in mice to define their ability to reach the brain. RESULTS: Brain uptake of OX26-conjugated NPs is much higher than of unmodified NPs, because: long-circulating abilities (conferred by PEG), interaction between cationic Cs and brain endothelium negative charges and OX26 TfR receptor affinity. CONCLUSION: Cs-PEG-OX26 NPs are promising drug delivery system to the CNS.


Subject(s)
Central Nervous System/drug effects , Chitosan/administration & dosage , Drug Delivery Systems , Nanoparticles/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Blood-Brain Barrier/drug effects , Brain/drug effects , Chitosan/adverse effects , Humans , Mice , Nanoparticles/adverse effects , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects
20.
J Liposome Res ; 25(2): 150-6, 2015.
Article in English | MEDLINE | ID: mdl-25203607

ABSTRACT

The development of smart delivery systems able to deliver and target a drug to the site of action is one of the major challenges in the field of pharmaceutical technology. The surface modification of nanocarriers, such as liposomes, is widely investigated either for increasing the blood circulation time (by pegylation) or for interacting with specific tissues or cells (by conjugation of a selective ligand as a monoclonal antibody, mAb). Microscopical analysis thereby is a useful approach to evaluate the morphology and the size owing to resolution and versatility in defining either surface modification or the architecture and the internal structure of liposomes. This contribution aims to connect the outputs obtained by transmission electron (TEM) and atomic force (AFM) microscopical techniques for identifying the modifications on the liposomal surface. To reach this objective, we prepared liposomes applying two different pegylation technologies and further modifying the surface by mAb conjugation. This work demonstrates the feasibility to apply the combined approach (TEM and AFM analysis) in the evaluation of the efficacy of a surface engineering process.


Subject(s)
Liposomes/chemistry , Microscopy, Atomic Force , Particle Size , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...