Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-37157144

ABSTRACT

Neurodegenerative diseases (NDs) such as Alzheimer's disease (AD) and Parkinson's disease (PD) are an accelerating global health problem as life expectancy rises worldwide. Despite their significant burden in public health systems to date, the existing treatments only manage the symptoms without slowing down disease progression. Thus, the ongoing neurodegenerative process remains untreated. Moreover, the stronghold of the brain-the blood-brain barrier (BBB)-prevents drug penetrance and dwindles effective treatments. In the last years, nanotechnology-based drug delivery systems (DDS) have become a promising approach to target and treat these disorders related to the central nervous system (CNS). PLGA based nanoparticles (NPs) were the first employed DDS for effective drug delivery. However, the poor drug loading capacity and localized immunogenicity prompted the scientific community to move to another DDS such as lipid-based NPs. Despite the lipid NPs' safety and effectiveness, their off-target accumulation together with the denominated CARPA (complement activation-related pseudo allergy) reaction has limited their complete clinical translation. Recently, biological NPs naturally secreted by cells, termed as extracellular vesicles (EVs) have emerged as promising more complex biocompatible DDS. In addition, EVs act as dual players in NDs treatment, as a "cell free" therapy themselves, as well as new biological NPs with numerous characteristics that qualify them as promising carriers over synthetic DDS. The present review aims to display advantages, drawbacks, current limitations and future prospective of the previously cited synthetic and biological DDS to enter the brain and treat one of 21st century most challenging diseases, NDs. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.


Subject(s)
Alzheimer Disease , Extracellular Vesicles , Nanoparticles , Parkinson Disease , Humans , Parkinson Disease/drug therapy , Alzheimer Disease/drug therapy , Brain , Blood-Brain Barrier , Drug Delivery Systems , Nanoparticles/therapeutic use , Lipids
2.
Cytotherapy ; 25(10): 1027-1032, 2023 10.
Article in English | MEDLINE | ID: mdl-37140499

ABSTRACT

BACKGROUND AIMS: Despite intensive research, to date, there is no effective treatment for neurodegenerative diseases. Among the different therapeutic approaches, recently, the use of extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) has gained attention. METHODS: In the present work, we focused on medium/large extracellular vesicles (m/lEVs) derived from hair follicle--derived (HF) MSCs, comparing their potential neuroprotective and anti-inflammatory effect against adipose tissue (AT)-MSC-derived m/lEVs. RESULTS: The obtained m/lEVs were similar in size with comparable expression of surface protein markers. The neuroprotective effect of both HF-m/lEVs and AT-m/lEVs was statistically significant in dopaminergic primary cell cultures, increasing cell viability after the incubation with 6-hidroxydopamine neurotoxin. Moreover, the administration of HF-m/lEVs and AT-m/lEVs counteracted the lipopolysaccharide-induced inflammation in primary microglial cell cultures, decreasing the levels of pro-inflammatory cytokines, tumor necrosis factor-α and interleukin-1ß. CONCLUSIONS: Taken together, HF-m/lEVs demonstrated comparable potential with that of AT-m/lEVs as multifaceted biopharmaceuticals for neurodegenerative disease treatment.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Neurodegenerative Diseases , Humans , Microglia , Hair Follicle , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Extracellular Vesicles/metabolism
3.
Fluids Barriers CNS ; 19(1): 22, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35300705

ABSTRACT

BACKGROUND: Neurodegenerative diseases (NDs) are an accelerating global health problem. Nevertheless, the stronghold of the brain- the blood-brain barrier (BBB) prevents drug penetrance and dwindles effective treatments. Therefore, it is crucial to identify Trojan horse-like drug carriers that can effectively cross the blood-brain barrier and reach the brain tissue. We have previously developed polyunsaturated fatty acids (PUFA)-based nanostructured lipid carriers (NLC), namely DHAH-NLC. These carriers are modulated with BBB-permeating compounds such as chitosan (CS) and trans-activating transcriptional activator (TAT) from HIV-1 that can entrap neurotrophic factors (NTF) serving as nanocarriers for NDs treatment. Moreover, microglia are suggested as a key causative factor of the undergoing neuroinflammation of NDs. In this work, we used in vitro models to investigate whether DHAH-NLCs can enter the brain via the BBB and investigate the therapeutic effect of NTF-containing DHAH-NLC and DHAH-NLC itself on lipopolysaccharide-challenged microglia. METHODS: We employed human induced pluripotent stem cell-derived brain microvascular endothelial cells (BMECs) to capitalize on the in vivo-like TEER of this BBB model and quantitatively assessed the permeability of DHAH-NLCs. We also used the HMC3 microglia cell line to assess the therapeutic effect of NTF-containing DHAH-NLC upon LPS challenge. RESULTS: TAT-functionalized DHAH-NLCs successfully crossed the in vitro BBB model, which exhibited high transendothelial electrical resistance (TEER) values (≈3000 Ω*cm2). Specifically, the TAT-functionalized DHAH-NLCs showed a permeability of up to 0.4% of the dose. Furthermore, using human microglia (HMC3), we demonstrate that DHAH-NLCs successfully counteracted the inflammatory response in our cultures after LPS challenge. Moreover, the encapsulation of glial cell-derived neurotrophic factor (GNDF)-containing DHAH-NLCs (DHAH-NLC-GNDF) activated the Nrf2/HO-1 pathway, suggesting the triggering of the endogenous anti-oxidative system present in microglia. CONCLUSIONS: Overall, this work shows that the TAT-functionalized DHAH-NLCs can cross the BBB, modulate immune responses, and serve as cargo carriers for growth factors; thus, constituting an attractive and promising novel drug delivery approach for the transport of therapeutics through the BBB into the brain.


Subject(s)
Blood-Brain Barrier , Nanoparticles , Nerve Growth Factors , Neurodegenerative Diseases , tat Gene Products, Human Immunodeficiency Virus , Blood-Brain Barrier/metabolism , Docosahexaenoic Acids/chemistry , Humans , Induced Pluripotent Stem Cells/metabolism , Liposomes , Microglia/metabolism , Nerve Growth Factors/administration & dosage , Neurodegenerative Diseases/drug therapy , tat Gene Products, Human Immunodeficiency Virus/chemistry
4.
Pharmaceutics ; 12(10)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003360

ABSTRACT

Neurodegenerative diseases (ND) are one of the main problems of public health systems in the 21st century. The rise of nanotechnology-based drug delivery systems (DDS) has become in an emerging approach to target and treat these disorders related to the central nervous system (CNS). Among others, the use of nanostructured lipid carriers (NLCs) has increased in the last few years. Up to today, most of the developed NLCs have been made of a mixture of solid and liquid lipids without any active role in preventing or treating diseases. In this study, we successfully developed NLCs made of a functional lipid, such as the hydroxylated derivate of docohexaenoic acid (DHAH), named DHAH-NLCs. The newly developed nanocarriers were around 100 nm in size, with a polydispersity index (PDI) value of <0.3, and they exhibited positive zeta potential due to the successful chitosan (CS) and TAT coating. DHAH-NLCs were shown to be safe in both dopaminergic and microglia primary cell cultures. Moreover, they exhibited neuroprotective effects in dopaminergic neuron cell cultures after exposition to 6-hydroxydopamine hydrochloride (6-OHDA) neurotoxin and decreased the proinflammatory cytokine levels in microglia primary cell cultures after lipopolysaccharide (LPS) stimuli. The levels of the three tested cytokines, IL-6, IL-1ß and TNF-α were decreased almost to control levels after the treatment with DHAH-NLCs. Taken together, these data suggest the suitability of DHAH-NLCs to attaining enhanced and synergistic effects for the treatment of NDs.

5.
Neurobiol Dis ; 121: 252-262, 2019 01.
Article in English | MEDLINE | ID: mdl-30296616

ABSTRACT

Omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been widely associated to beneficial effect over different neurodegenerative diseases. In the present study, we tested the potential therapeutic effect of docohexanoic acid (DHA) and its hydroxylated derivate, DHAH, in a partial lesion model of Parkinson's disease (PD). One month before and four months after the striatal lesion with 6-OHDA was made, the animals were daily treated with DHA (50 mg/kg), DHAH (50 mg/kg), vehicle or saline, by intragastric administration. Animal groups under n-3 PUFA treatments exhibited a trend to improve in amphetamine-induced rotations and cylinder test. The beneficial effect seen in behavioral studies were confirmed with TH immunostaining. TH+ fibers and TH+ neurons increased in the experimental groups treated with both n-3 PUFAs, DHA and DHAH. Moreover, the n-3 PUFAs administration decreased the astrogliosis and microgliosis, in both the striatum and substantia nigra (SN), with a higher decrease of GFAP+ and Iba-1+ cells for the DHAH treated group. This experimental group also revealed a positive effect on Nrf2 pathway regulation, decreasing the positive Nrf2 immmunostaining in the striatum and SN, which revealed a potential antioxidant effect of this compound. Taking together, these data suggest a positive effect of n-3 PUFAs administration, and more concretely of DHAH, for PD treatment as it exhibited positive results on dopaminergic system, neuroinflammation and oxidative stress.


Subject(s)
Corpus Striatum/drug effects , Dopaminergic Neurons/drug effects , Fatty Acids, Omega-3/administration & dosage , Neuroglia/drug effects , Oxidative Stress/drug effects , Parkinson Disease/metabolism , Amphetamine/administration & dosage , Animals , Antioxidants/administration & dosage , Corpus Striatum/metabolism , Dopamine/metabolism , Dopamine Agents/administration & dosage , Dopaminergic Neurons/metabolism , Male , Motor Activity/drug effects , NF-E2-Related Factor 2/metabolism , Neuroglia/metabolism , Oxidopamine/administration & dosage , Parkinson Disease/prevention & control , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
7.
Mol Neurobiol ; 55(1): 145-155, 2018 01.
Article in English | MEDLINE | ID: mdl-28866799

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder (ND), characterized by the loss of dopaminergic neurons, microglial activation, and neuroinflammation. Current available treatments in clinical practice cannot halt the progression of the disease. During the last few years, growth factors (GFs) have been raised as a promising therapeutic approach to address the underlying neurodegenerative process. Among others, glial cell-derived neurotrophic factor (GDNF) is a widely studied GF for PD. However, its clinical use is limited due to its short half life, rapid degradation rate, and difficulties in crossing the blood-brain barrier (BBB). Lately, intranasal administration has appeared as an alternative non-invasive way to bypass the BBB and target drugs directly to the central nervous system (CNS). Thus, the aim of this work was to develop a novel nanoformulation to enhance brain targeting in PD through nasal administration. For that purpose, GDNF was encapsulated into chitosan (CS)-coated nanostructured lipid carriers, with the surface modified with transactivator of transcription (TAT) peptide (CS-nanostructured lipid carrier (NLC)-TAT-GDNF). After the physiochemical characterization of nanoparticles, the in vivo study was performed by intranasal administration to a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. The CS-NLC-TAT-GDNF-treated group revealed motor recovery which was confirmed with immunohistochemistry studies, showing the highest number of tyrosine hydroxylase (TH+) fibers in the striatum and TH+ neuron levels in the substantia nigra. Moreover, ionizing calcium-binding adaptor molecule 1 immunohistochemistry was performed, revealing that CS-NLC-TAT-GDNF acts as a modulator on microglia activation, obtaining values similar to control. Therefore, it may be concluded that the intranasal administration of CS-NLC-TAT-GDNF may represent a promising therapy for PD treatment.


Subject(s)
Drug Carriers/administration & dosage , Gene Products, tat/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Nanoparticles/administration & dosage , Parkinsonian Disorders/drug therapy , Administration, Intranasal , Animals , Lipids , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/pathology
8.
Nanomedicine (Lond) ; 11(10): 1267-85, 2016 05.
Article in English | MEDLINE | ID: mdl-27077453

ABSTRACT

Alzheimer's disease and Parkinson's disease are the most common neurodegenerative diseases worldwide. Despite all the efforts made by the scientific community, current available treatments have limited effectiveness, without halting the progression of the disease. That is why, new molecules such as growth factors, antioxidants and metal chelators have been raised as new therapeutical approaches. However, these molecules have difficulties to cross the blood-brain barrier limiting its therapeutic effect. The development of nanometric drug delivery systems may permit a targeted and sustained release of old and new treatments offering a novel strategy to treat these neurodegenerative disorders. This review summarized the main investigated drug delivery systems as promising approaches to treat Alzheimer's disease and Parkinson's disease.


Subject(s)
Alzheimer Disease/drug therapy , Drug Delivery Systems/methods , Nanomedicine/methods , Parkinson Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Discovery , Humans , Nanoparticles/chemistry , Nanoparticles/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/therapeutic use , Parkinson Disease/metabolism , Parkinson Disease/physiopathology
9.
Pest Manag Sci ; 72(4): 707-18, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25989043

ABSTRACT

BACKGROUND: Sulfoxaflor, a new insecticide from the sulfoximine chemical family, and imidacloprid, a widely used neonicotinoid insecticide, were tested to assess the susceptibility and feeding behaviour of two populations of Myzus persicae: Mp61, which exhibited target-site R81T resistance to neonicotinoids, and Mp1989, a laboratory clone maintained since 1989 as a susceptible reference. RESULTS: The imidacloprid LC50 value for Mp61 was 16 times higher than for Mp1989, showing a moderate level of resistance. Sulfoxaflor LC50 values for Mp61 and Mp1989 were much closer. The probing behaviour, as assessed by electrical penetration graphs (EPGs), of both populations was clearly altered by sulfoxaflor, which reduced the ability of aphids to find and feed from the phloem. The feeding behaviour of the susceptible Mp1989 population was much more severely affected than the moderately resistant Mp61 population on imidacloprid-treated plants. PCR assays of both aphid populations followed by DNA sequencing identified differences between populations in the point mutation in the ß-subunit of the nicotinic acetylcholine receptor linked to the resistant gene against the neonicotinoid insecticide. CONCLUSIONS: Sulfoxaflor provoked feeding cessation more rapidly than imidacloprid in both aphid populations. Sharp differences in feeding behaviour were detected between the susceptible and the moderately resistant neonicotinoid-resistant aphid populations. The EPG technique can be used as a useful tool to give new insights into the functional effects of new chemical compounds and for early detection of low to moderate levels of resistance of sap-feeding insects to insecticides. The potential of this technique was validated by molecular analysis of the R81T mutation target site.


Subject(s)
Aphids/drug effects , Biological Assay/methods , Electricity , Insecticides/pharmacology , Animals , Aphids/genetics , Aphids/physiology , DNA Mutational Analysis , Feeding Behavior/drug effects , Imidazoles/pharmacology , Insecticide Resistance/genetics , Neonicotinoids , Nitro Compounds/pharmacology , Pyridines/pharmacology , Sulfur Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...