Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Biol Chem ; 295(33): 11866-11876, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32616652

ABSTRACT

Garcinoic acid (GA or δ-T3-13'COOH), is a natural vitamin E metabolite that has preliminarily been identified as a modulator of nuclear receptors involved in ß-amyloid (Aß) metabolism and progression of Alzheimer's disease (AD). In this study, we investigated GA's effects on Aß oligomer formation and deposition. Specifically, we compared them with those of other vitamin E analogs and the soy isoflavone genistein, a natural agonist of peroxisome proliferator-activated receptor γ (PPARγ) that has therapeutic potential for managing AD. GA significantly reduced Aß aggregation and accumulation in mouse cortical astrocytes. Similarly to genistein, GA up-regulated PPARγ expression and apolipoprotein E (ApoE) efflux in these cells with an efficacy that was comparable with that of its metabolic precursor δ-tocotrienol and higher than those of α-tocopherol metabolites. Unlike for genistein and the other vitamin E compounds, the GA-induced restoration of ApoE efflux was not affected by pharmacological inhibition of PPARγ activity, and specific activation of pregnane X receptor (PXR) was observed together with ApoE and multidrug resistance protein 1 (MDR1) membrane transporter up-regulation in both the mouse astrocytes and brain tissue. These effects of GA were associated with reduced Aß deposition in the brain of TgCRND8 mice, a transgenic AD model. In conclusion, GA holds potential for preventing Aß oligomerization and deposition in the brain. The mechanistic aspects of GA's properties appear to be distinct from those of other vitamin E metabolites and of genistein.


Subject(s)
Amyloid beta-Peptides/metabolism , Benzopyrans/pharmacology , Brain/drug effects , Protein Aggregation, Pathological/prevention & control , Vitamin E/analogs & derivatives , Amyloid beta-Peptides/ultrastructure , Animals , Benzopyrans/pharmacokinetics , Brain/metabolism , Brain/pathology , Male , Mice , Protein Aggregates/drug effects , Protein Aggregation, Pathological/pathology , Vitamin E/pharmacokinetics , Vitamin E/pharmacology
2.
Int J Mol Sci ; 20(19)2019 Sep 24.
Article in English | MEDLINE | ID: mdl-31554165

ABSTRACT

The increase in the incidence of neurodegenerative diseases, in particular Alzheimer's Disease (AD), is a consequence of the world's population aging but unfortunately, existing treatments are only effective at delaying some of the symptoms and for a limited time. Despite huge efforts by both academic researchers and pharmaceutical companies, no disease-modifying drugs have been brought to the market in the last decades. Recently, several studies shed light on Carbonic Anhydrases (CAs, EC 4.2.1.1) as possible new targets for AD treatment. In the present review we summarized preclinical and clinical findings regarding the role of CAs and their inhibitors/activators on cognition, aging and neurodegeneration and we discuss future challenges and opportunities in the field.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Carbonic Anhydrases/metabolism , Alzheimer Disease/drug therapy , Animals , Biomarkers , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/therapeutic use , Carbonic Anhydrases/genetics , Central Nervous System/drug effects , Central Nervous System/metabolism , Disease Models, Animal , Disease Susceptibility , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Humans , Molecular Targeted Therapy , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/metabolism , Oxidative Stress/drug effects
3.
Br J Clin Pharmacol ; 83(1): 54-62, 2017 01.
Article in English | MEDLINE | ID: mdl-27131215

ABSTRACT

AIM: In TgCRND8 (Tg) mice we checked the dose-response effect of diet supplementation with oleuropein aglycone (OLE) at 12.5 or 0.5 mg kg-1 of diet. We also studied the effects of dietary intake of the mix of polyphenols present in olive mill waste water administered at a total dose as high as the highest dose of OLE (50 mg kg-1 of diet) previously investigated. METHODS: Four month-old Tg mice were equally divided into four groups and treated for 8 weeks with a modified low fat (5.0%) AIN-76 A diet (10 g day-1  per mouse) as such, supplemented with OLE (12.5 or 0.5 mg kg-1 of diet) or with a mix of polyphenols (50 mg kg-1 of diet) found in olive mill waste water. Behavioural performance was evaluated by the step down inhibitory avoidance and object recognition tests. Neuropathology was analyzed by immunohistochemistry. RESULTS: OLE supplementation at 12.5 mg kg-1 of diet and the mix of polyphenols was found to improve significantly cognitive functions of Tg mice (P < 0.0001). Aß42 and pE-3Aß plaque area and number were significantly reduced in the cortex by OLE and in the cortex and hippocampus by the mix of polyphenols (P < 0.01, P < 0.001 and P < 0.0001). Similar autophagy induction was found in the brain cortex of differently treated mice. CONCLUSION: Our results extend previous data showing that the effects of OLE on behavioural performance and neuropathology are dose-dependent and not closely related to OLE by itself. In fact, diet supplementation with the same dose of a mix of polyphenols found in olive mill waste water resulted in comparable neuroprotection.


Subject(s)
Brain/drug effects , Cognitive Dysfunction/drug therapy , Iridoids/therapeutic use , Olea/chemistry , Plaque, Amyloid/drug therapy , Polyphenols/therapeutic use , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Dietary Supplements , Disease Models, Animal , Dose-Response Relationship, Drug , Iridoid Glucosides , Iridoids/isolation & purification , Mice, Transgenic , Peptide Fragments/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Polyphenols/isolation & purification , Wastewater/chemistry
4.
Planta Med ; 83(5): 382-391, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27002395

ABSTRACT

The blood-brain barrier hinders the passage of systemically delivered therapeutics and the brain extracellular matrix limits the distribution and durability of locally delivered agents. Drug-loaded nanocarriers represent a promising strategy to overcome these barriers and address specific drug delivery challenges due to their small size and versatile design. We synthetized [fluorescent poly(ethyl-cyanoacrylate) nanoparticles coated with Tween 80 by an emulsion polymerization method to target and reach the brain after intravenous and intraperitoneal administration. Nanoparticles were characterized in terms of dimensional analysis, polydispersity and zeta potential (ζ-potential), morphology, encapsulation efficacy, and loading capacity. After intracerebral injection in healthy rats, nanoparticles were distributed within the injected hemisphere and mainly interacted with microglial cells, presumably involved in their clearance by phagocytosis. Furthermore, nanoparticles were able to pass the blood-brain barrier after systemic administration in rats, and the lack of toxicity in C57/B6 mice chronically administered was highlighted. The data obtained helped to clarify the nanoparticles distribution, accumulation, fate, and toxicity into the brain. The selected nanoparticles may represent a biocompatible promising carrier to be further investigated as brain delivery systems. Salvianolic acid B from Salvia miltiorrhiza is a promising molecule in the protection of degeneration in several animal models by various biological mechanisms, but its poor chemical stability and low bioavailability limits its clinical application for central nervous system neuronal injury and degeneration. Nanoparticles were loaded with salvianolic acid B obtaining an encapsulation efficacy and loading capacities of 98.70 % ± 0.45 and 53.3 % ± 0.24, respectively. They were suitable for parental administration because their mean diameter was smaller than 300 nm, with a polydispersity of 0.04 ± 0.03, and a ζ-potential of - 8.38 mV ± 3.87. The in vitro release of salvianolic acid B from the nanoparticles was sustained and prolonged during 8 h, suitable for a promising clinical application.


Subject(s)
Benzofurans/administration & dosage , Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Drug Carriers/metabolism , Nanoparticles/metabolism , Animals , Biocompatible Materials , Cyanoacrylates , Drug Carriers/chemistry , Female , Male , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Rats , Rats, Wistar , Salvia miltiorrhiza/chemistry
5.
Biochim Biophys Acta ; 1832(8): 1217-26, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23602994

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterised by cognitive decline, formation of the extracellular amyloid ß (Aß42) plaques, neuronal and synapse loss, and activated microglia and astrocytes. Extracellular chaperones, which are known to inhibit amyloid fibril formation and promote clearance of misfolded aggregates, have recently been shown to reduce efficiently the toxicity of HypF-N misfolded oligomers to immortalised cell lines, by binding and clustering them into large species. However, the role of extracellular chaperones on Aß oligomer toxicity remains unclear, with reports often appearing contradictory. In this study we microinjected into the hippocampus of rat brains Aß42 oligomers pre-incubated for 1h with two extracellular chaperones, namely clusterin and α2-macroglobulin. The chaperones were found to prevent Aß42-induced learning and memory impairments, as assessed by the Morris Water Maze test, and reduce Aß42-induced glia inflammation and neuronal degeneration in rat brains, as probed by fluorescent immunohistochemical analyses. Moreover, the chaperones were able to prevent Aß42 colocalisation with PSD-95 at post-synaptic terminals of rat primary neurons, suppressing oligomer cytotoxicity. All such effects were not effective by adding pre-formed oligomers and chaperones without preincubation. Molecular chaperones have therefore the potential to prevent the early symptoms of AD, not just by inhibiting Aß42 aggregation, as previously demonstrated, but also by suppressing the toxicity of Aß42 oligomers after they are formed. These findings elect them as novel neuroprotectors against amyloid-induced injury and excellent candidates for the design of therapeutic strategies against AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Molecular Chaperones/metabolism , Peptide Fragments/metabolism , Animals , Cells, Cultured , Inflammation/metabolism , Learning Disabilities/metabolism , Male , Memory Disorders/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , alpha-Macroglobulins/metabolism
6.
Neurodegener Dis ; 13(2-3): 131-4, 2014.
Article in English | MEDLINE | ID: mdl-24192327

ABSTRACT

BACKGROUND: Translational research needs valid animal models of disease to discover new pathogenetic aspects and treatments. In Alzheimer's disease (AD), transgenic models are of great value for AD research and drug testing. OBJECTIVE: It was the aim of this study to analyze the power of dietary polyphenols against neurodegeneration by investigating the effects of oleuropein aglycone (OLE), the main phenol in the extra virgin olive oil (EVOO), a key component of the Mediterranean diet (MD), in a mouse model of amyloid-ß deposition. METHODS: TgCRND8 mice (3.5 months old), expressing the mutant KM670/671NL+V717F h-ßAPP695 transgene, and wild-type (wt) mice were used to study in vivo the effects of an 8-week dietary supplementation with OLE (50 mg/kg of diet) [Grossi et al: PLoS One 2013;8:e71702], following the European Communities Council Directive 86/609 (DL 116/92) and National Guidelines (permit number: 283/2012-B). RESULTS: OLE administration ameliorates memory dysfunction, raises a significant autophagic response in the cortex and promotes the proliferation of newborn cells in the subgranular zone of the dentate gyrus of the hippocampus. CONCLUSIONS: Our findings support the beneficial effects of EVOO and highlight the possibility that continuous intake of high doses of OLE, both as a nutraceutical or as a food integrator, may prevent/delay the appearance of AD and reduce the severity of its symptoms.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Diet, Mediterranean , Iridoids/pharmacology , Neuroprotective Agents/pharmacology , Animal Feed , Animals , Disease Models, Animal , Humans , Iridoid Glucosides , Mice, Transgenic , Olive Oil , Plant Oils/chemistry , Translational Research, Biomedical
7.
Cells ; 12(18)2023 09 12.
Article in English | MEDLINE | ID: mdl-37759482

ABSTRACT

The term neuroinflammation defines the reactions of astrocytes and microglia to alterations in homeostasis in the diseased central nervous system (CNS), the exacerbation of which contributes to the neurodegenerative effects of Alzheimer's disease (AD). Local environmental conditions, such as the presence of proinflammatory molecules, mechanical properties of the extracellular matrix (ECM), and local cell-cell interactions, are determinants of glial cell phenotypes. In AD, the load of the cytotoxic/proinflammatory amyloid ß (Aß) peptide is a microenvironmental component increasingly growing in the CNS, imposing time-evolving challenges on resident cells. This study aimed to investigate the temporal and spatial variations of the effects produced by this process on astrocytes and microglia, either directly or by interfering in their interactions. Ex vivo confocal analyses of hippocampal sections from the mouse model TgCRND8 at different ages have shown that overproduction of Aß peptide induced early and time-persistent disassembly of functional astroglial syncytium and promoted a senile phenotype of reactive microglia, hindering Aß clearance. In the late stages of the disease, these patterns were altered in the presence of Aß-plaques, surrounded by typically reactive astrocytes and microglia. Morphofunctional characterization of peri-plaque gliosis revealed a direct contribution of astrocytes in plaque buildup that might result in shielding Aß-peptide cytotoxicity and, as a side effect, in exacerbating neuroinflammation.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/genetics , Amyloid beta-Peptides , Mice, Transgenic , Astrocytes , Neuroinflammatory Diseases , Central Nervous System , Plaque, Amyloid
8.
Antioxidants (Basel) ; 11(10)2022 Sep 21.
Article in English | MEDLINE | ID: mdl-36290586

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease representing the most prevalent cause of dementia. It is also related to the aberrant amyloid-beta (Aß) protein deposition in the brain. Since oxidative stress is involved in AD, there is a possible role of antioxidants present in the effected person's diet. Thus, we assessed the effect of the systemic administration of solid lipid nanoparticles (SLNs) to facilitate curcumin (CUR) delivery on TG2 isoform expression levels in Wild Type (WT) and in TgCRND8 (Tg) mice. An experimental model of AD, which expresses two mutated human amyloid precursor protein (APP) genes, was used. Behavioral studies were also performed to evaluate the improvement of cognitive performance and memory function induced by all treatments. The expression levels of Bcl-2, Cyclin-D1, and caspase-3 cleavage were evaluated as well. In this research, for the first time, we demonstrated that the systemic administration of SLNs-CUR, both in WT and in Tg mice, allows one to differently modulate TG2 isoforms, which act either on apoptotic pathway activation or on the ability of the protein to repair cellular damage in the brains of Tg mice. In this study, we also suggest that SLNs-CUR could be an innovative tool for the treatment of AD.

9.
J Cell Mol Med ; 15(10): 2106-16, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21155974

ABSTRACT

Peptides and proteins can convert from their soluble forms into highly ordered fibrillar aggregates, giving rise to pathological conditions ranging from neurodegenerative disorders to systemic amyloidoses. It is increasingly recognized that protein oligomers forming early in the process of fibril aggregation represent the pathogenic species in protein deposition diseases. The N-terminal domain of the HypF protein from Escherichia coli (HypF-N) has previously been shown to form, under distinct conditions, two types of HypF-N oligomers with indistinguishable morphologies but distinct structural features at the molecular level. Only the oligomer type exposing hydrophobic surfaces and possessing sufficient structural plasticity is toxic (type A), whereas the other type is benign to cultured cells (type B). Here we show that only type A oligomers are able to induce a Ca(2+) influx from the cell medium to the cytosol, to penetrate the plasma membrane, to increase intracellular reactive oxygen species production, lipid peroxidation and release of intracellular calcein, resulting in the activation of the apoptotic pathway. Remarkably, these oligomers can also induce a loss of cholinergic neurons when injected into rat brains. By contrast, markers of cellular stress and viability were unaffected in cultured and rat neuronal cells exposed to type B oligomers. The analysis of the time scales of such effects indicates that the difference of toxicity between the two oligomer types involve the early events of the toxicity cascade, shedding new light on the mechanism of action of protein oligomers and on the molecular targets for the therapeutic intervention against protein deposition diseases.


Subject(s)
Calcium/metabolism , Carboxyl and Carbamoyl Transferases/chemistry , Carboxyl and Carbamoyl Transferases/pharmacology , Cholinergic Neurons/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/pharmacology , Peptides/chemistry , Peptides/pharmacology , Amyloidogenic Proteins/chemistry , Amyloidogenic Proteins/metabolism , Animals , Apoptosis/drug effects , Cell Membrane Permeability/drug effects , Cells, Cultured , Cholinergic Neurons/chemistry , Disease Models, Animal , Humans , Lipid Peroxidation/drug effects , Molecular Targeted Therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
10.
Antioxidants (Basel) ; 10(7)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209636

ABSTRACT

Oxidative stress and inflammation triggered by increased oxidative stress are the cause of many chronic diseases. The lack of anti-inflammatory drugs without side-effects has stimulated the search for new active substances. Plant-derived compounds provide new potential anti-inflammatory and antioxidant molecules. Natural products are structurally optimized by evolution to serve particular biological functions, including the regulation of endogenous defense mechanisms and interaction with other organisms. This property explains their relevance for infectious diseases and cancer. Recently, among the various natural substances, polyphenols from extra virgin olive oil (EVOO), an important element of the Mediterranean diet, have aroused growing interest. Extensive studies have shown the potent therapeutic effects of these bioactive molecules against a series of chronic diseases, such as cardiovascular diseases, diabetes, neurodegenerative disorders and cancer. This review begins from the chemical structure, abundance and bioavailability of the main EVOO polyphenols to highlight the effects and the possible molecular mechanism(s) of action of these compounds against inflammation and oxidation, in vitro and in vivo. In addition, the mechanisms of inhibition of molecular signaling pathways activated by oxidative stress by EVOO polyphenols are discussed, together with their possible roles in inflammation-mediated chronic disorders, also taking into account meta-analysis of population studies and clinical trials.

11.
J Neurochem ; 112(6): 1539-51, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20050968

ABSTRACT

To investigate the role of the Wnt inhibitor Dickkopf-1 (DKK-1) in the pathophysiology of neurodegenerative diseases, we analysed DKK-1 expression and localization in transgenic mouse models expressing familial Alzheimer's disease mutations and a frontotemporal dementia mutation. A significant increase of DKK-1 expression was found in the diseased brain areas of all transgenic lines, where it co-localized with hyperphosphorylated tau-bearing neurons. In TgCRND8 mice, DKK-1 immunoreactivity was detected in neurons surrounding amyloid deposits and within the choline acetyltransferase-positive neurons of the basal forebrain. Active glycogen synthase kinase-3 (GSK-3) was found to co-localize with DKK-1 and phospho-tau staining. Downstream to GSK-3, a significant reduction in beta-catenin translocation to the nucleus, indicative of impaired Wnt signaling functions, was found as well. Cumulatively, our findings indicate that DKK-1 expression is associated with events that lead to neuronal death in neurodegenerative diseases and support a role for DKK-1 as a key mediator of neurodegeneration with therapeutic potential.


Subject(s)
Gene Expression Regulation/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Neurodegenerative Diseases/metabolism , Age Factors , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Antibodies, Monoclonal/metabolism , Brain/cytology , Brain/metabolism , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Gene Expression Regulation/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurons/metabolism , Peptide Fragments/metabolism , Phosphopyruvate Hydratase/metabolism , Presenilin-1/genetics , beta Catenin/metabolism , tau Proteins/metabolism
12.
Mol Immunol ; 45(4): 1056-62, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17850871

ABSTRACT

In vitro and in vivo studies indicate that Alzheimer's Disease (AD) could be prevented or treated by active immunization against self-peptide beta-amyloid. In this study, we compared the immunogenicity of different regions of beta-amyloid, displayed on filamentous phages. We established that a filamentous phage displaying epitope 2-6 (AEFRH) of beta-amyloid at the N-terminus of Major Capside Protein (phage fdAD(2-6)) is more immunogenic than a phage displaying epitope 1-7 (DAEFRHD) that differs only in flanking residues. Monthly injections of fdAD(2-6) trigger a robust anti-beta-amyloid antibody response, and afford a significant reduction of plaque pathology in a mouse model of AD, whereas the same treatment, performed with phage fdAD(1-7), induces a lower anti-beta-amyloid titer and does not protect from amyloid deposition. "Memory" anti-amyloid antibodies induced by a single prime-boost cycle with vaccine fdAD(2-6), that have a lower titer compared to antibodies induced by monthly restimulations, do not prevent plaque pathology. Our data show that optimization of epitope display is essential in vaccine design, and suggest that the titer of the anti-amyloid response is the crucial parameter to obtain therapeutic efficacy in vivo.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Antibodies/immunology , Coliphages/immunology , Peptide Fragments/immunology , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Animals , Antibodies/therapeutic use , Coliphages/metabolism , Epitopes , Immunotherapy , Mice , Mice, Transgenic , Peptide Fragments/therapeutic use , Peptide Library , Plaque, Amyloid/pathology
13.
Front Pharmacol ; 10: 910, 2019.
Article in English | MEDLINE | ID: mdl-31507412

ABSTRACT

Andrographolide (AG) was encapsulated in human albumin nanoparticles (AG NPs), and their crossing properties of the blood-brain barrier (BBB), brain distribution, and effects in TgCRND8 mice were evaluated. The development of appropriate NP formulations is mandatory because of the scarce BBB permeability properties of AG. Developed NPs had proper size (mean size: 159.2 ± 4.5 nm), size distribution (PDI nearby 0.12 ± 0.01), and ζ potential (-24.8 ± 1.2 mV), which were not affected by sodium fluorescein (NAF) loading. When AG was loaded to NPs, it slightly affected their size (210.4 ± 3.2 nm) and ζ potential (-20.3 ± 1.5) but not the PDI. Both NAF and AG had a remarkable encapsulation efficiency (more than 99%). The in vitro release of AG from the NPs reached the highest percentage (48%) after 24 h, and the Higuchi's equation was found to be the best fitting model (R2 = 0.9635). Both AG and AG NPs did not alter the viability of N2a murine neuroblastoma cells when compared with the untreated control cells. In the step-down inhibitory avoidance test, AG NPs administered to TgCRND8 mice significantly improved their performance (P < 0.0001), reaching levels comparable to those displayed by wild-type mice. In the object recognition test, treated and untreated animals showed no deficiencies in exploratory activity, directional movement toward objects, and locomotor activity. No cognitive impairments (discrimination score) were detected in TgCRND8 mice (P < 0.0001) treated with AG NPs. After acute intravenous administration (200 µl), NPs loaded with the probe NAF were detected in the brain parenchyma of TgCRND8 mice. Immunofluorescent analyses evidenced the presence of NPs both in the pE3-Aß plaque surroundings and inside the pE3-Aß plaque, indicative of the ability of these NPs to cross the BBB and to penetrate in both undamaged and damaged brain tissues. Furthermore, the immunohistochemical analysis of GFAP-positive astrocytes in the hippocampus of Tg mice evidenced the anti-inflammatory activity of AG when AG NPs were intraperitoneally administered. AG was not effective in counteracting amyloid Aß aggregation and the resulting toxicity but significantly decreased the oxidative stress levels. In conclusion, AG NPs have extraordinary versatility, nontoxicity, nonimmunogenicity, strong biocompatibility, high biodegradability, and astonishing loading capacity of drug.

14.
Neurobiol Dis ; 31(1): 145-58, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18571100

ABSTRACT

In a comprehensive proteomics study aiming at the identification of proteins associated with amyloid-beta (Abeta)-mediated toxicity in cultured cortical neurons, we have identified Thimet oligopeptidase (THOP1). Functional modulation of THOP1 levels in primary cortical neurons demonstrated that its overexpression was neuroprotective against Abeta toxicity, while RNAi knockdown made neurons more vulnerable to amyloid peptide. In the TgCRND8 transgenic mouse model of amyloid plaque deposition, an age-dependent increase of THOP1 expression was found in brain tissue, where it co-localized with Abeta plaques. In accordance with these findings, THOP1 expression was significantly increased in human AD brain tissue as compared to non-demented controls. These results provide compelling evidence for a neuroprotective role of THOP1 against toxic effects of Abeta in the early stages of AD pathology, and suggest that the observed increase in THOP1 expression might be part of a compensatory defense mechanism of the brain against an increased Abeta load.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/toxicity , Cerebral Cortex/enzymology , Metalloendopeptidases/biosynthesis , Neurons/enzymology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Blotting, Western , Cells, Cultured , Cerebral Cortex/pathology , Female , Gene Expression , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Middle Aged , Neurons/pathology , Plaque, Amyloid/metabolism , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transfection
15.
J Alzheimers Dis ; 63(3): 1161-1172, 2018.
Article in English | MEDLINE | ID: mdl-29710709

ABSTRACT

Alzheimer's disease is the most common form of dementia affecting a large proportion of aged people. Plant polyphenols have been reported to be potentially useful in the prevention of AD due to their multiple pharmacological activities. The aim of the present study was to assess whether the previously reported neuroprotective and anti-inflammatory effects resulting from oleuropein aglycone administration were reproduced by diet supplementation with similar amounts of its metabolite hydoxytyrosol (HT). Four-month-old TgCRND8 and wild type mice were treated for 8 weeks with a low-fat diet (5%) supplemented with HT (50 mg/kg of diet). We found that HT supplementation significantly improved cognitive functions of TgCRND8 mice and significantly reduced Aß42 and pE3-Aß plaque area and number in the cortex; in the hippocampal areas of HT-fed TgCRND8 mice, we found a significant reduction in the pE3-Aß plaque number together with a tendency toward a reduction in Aß42 load and pE3-Aß plaque area, associated with a marked reduction of TNF-α expression and astrocyte reaction. Macroautophagy induction and modulation of MAPKs signaling were found to underlie the beneficial effects of HT. Our findings indicate that HT administration reproduces substantially the beneficial effects on behavioral performance and neuropathology previously reported in TgCRND8 mice fed with oleuropein aglycone, resulting in comparable neuroprotection.


Subject(s)
Alzheimer Disease/pathology , Antioxidants/therapeutic use , Brain/metabolism , Cognition Disorders/diet therapy , Cognition Disorders/etiology , Diet , Phenylethyl Alcohol/analogs & derivatives , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Amyloidosis, Familial/metabolism , Animals , Autophagy/drug effects , Brain/pathology , Corneal Dystrophies, Hereditary/metabolism , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Phenylethyl Alcohol/therapeutic use , Presenilin-1/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
16.
Front Aging Neurosci ; 10: 372, 2018.
Article in English | MEDLINE | ID: mdl-30483118

ABSTRACT

We investigated the different patterns of neurodegeneration and glia activation in CA1 and CA3 hippocampal areas of TgCRND8 mice. The main feature of this transgenic model is the rapid development of the amyloid pathology, which starts already at 3 months of age. We performed immunohistochemical analyses to compare the different sensibility of the two hippocampal regions to neurodegeneration. We performed qualitative and quantitative evaluations by fluorescence immunohistochemistry with double or triple staining, followed by confocal microscopy and digital image analysis in stratum pyramidale (SP) and stratum radiatum (SR) of CA1 and CA3, separately. We evaluated time-dependent Aß plaques deposition, expression of inflammatory markers, as well as quantitative and morphological alterations of neurons and glia in transgenic mice at 3 (Tg 3M) and 6 (Tg 6M) months of age, compared to WT mice. In CA1 SR of Tg 6M mice, we found significantly more Medium and Large plaques than in CA3. The pattern of neurodegeneration and astrocytes activation was different in the two areas, indicating higher sensitivity of CA1. In the CA1 SP of Tg 6M mice, we found signs of reactive astrogliosis, such as increase of astrocytes density in SP, increase of GFAP expression in SR, and elongation of astrocytes branches. We found also common patterns of glia activation and neurodegenerative processes in CA1 and CA3 of Tg 6M mice: significant increase of total and reactive microglia density in SP and SR, increased expression of TNFα, of iNOS, and IL1ß in astrocytes and increased density of neurons-astrocytes-microglia triads. In CA1 SP, we found decrease of volume and number of pyramidal neurons, paralleled by increase of apoptosis, and, consequently, shrinkage of CA1 SP. These data demonstrate that in TgCRND8 mice, the responses of neurons and glia to neurodegenerative patterns induced by Aß plaques deposition is not uniform in the two hippocampal areas, and in CA1 pyramidal neurons, the higher sensitivity may be related to the different plaque distribution in this area. All these modifications may be at the basis of memory loss, the peculiar symptom of AD, which was demonstrated in this transgenic mouse model of Aß deposition, even at early stages.

17.
Colloids Surf B Biointerfaces ; 161: 302-313, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29096375

ABSTRACT

Andrographolide is a major diterpenoid of Andrographis paniculata and possesses several biological activities, including protection against oxidative stress mediated neurotoxicity, inflammation-mediated neurodegeneration, and cerebral ischemia. However, this molecule shows low bioavailability, poor water solubility, and high chemical and metabolic instability. The present study reports preparation of solid lipid nanoparticles (SLN) to deliver andrographolide (AG) into the brain. SLN were prepared using Compritol 888 ATO as solid lipid and Brij 78 as surfactant and applying emulsion/evaporation/solidifying method as preparative procedure. Nanoparticles have a spherical shape, small dimensions, and narrow size distribution. Encapsulation efficiency of AG-loaded SLN was found to be 92%. Nanoparticles showed excellent physical and chemical stability during storage at 4°C for one month. The lyophilized product was also stable at 25°C during the same period. SLN remained unchanged also in the presence of human serum albumin and plasma. In vitro release at pH 7.4 was also studied. The release of AG was prolonged and sustained when the compound was entrapped in SLN. The ability of SLN to cross the blood-brain barrier (BBB) was evaluated first in vitro by applying a permeation test with artificial membrane (parallel artificial membrane permeability assay, PAMPA) to predict passive and transcellular permeability through the BBB, and then by using hCMEC/D3 cells, a well-established in vitro BBB model. In vitro results proved that nanoparticles improved permeability of AG compared to free AG. Fluorescent nanoparticles were then prepared for in vivo tests in healthy rats. After intravenous administration, fluorescent SLN were detected in brain parenchyma outside the vascular bed, confirming their ability to overcome the BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Diterpenes/administration & dosage , Drug Delivery Systems/methods , Lipids/chemistry , Nanoparticles/chemistry , Administration, Intravenous , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Brain/cytology , Brain/metabolism , Cell Line , Cell Survival/drug effects , Diterpenes/chemistry , Diterpenes/pharmacokinetics , Drug Liberation , Humans , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Rats, Wistar , Tissue Distribution
18.
J Neurosci ; 26(31): 8160-7, 2006 Aug 02.
Article in English | MEDLINE | ID: mdl-16885229

ABSTRACT

More than 40 human diseases are associated with fibrillar deposits of specific peptides or proteins in tissue. Amyloid fibrils, or their precursors, can be highly toxic to cells, suggesting their key role in disease pathogenesis. Proteins not associated with any disease are able to form oligomers and amyloid assemblies in vitro displaying structures and cytotoxicity comparable with those of aggregates of disease-related polypeptides. In isolated cells, such toxicity has been shown to result from increased membrane permeability with disruption of ion homeostasis and oxidative stress. Here we microinjected into the nucleus basalis magnocellularis of rat brains aggregates of an Src homology 3 domain and the N-terminal domain of the prokaryotic HypF, neither of which is associated with amyloid disease. Prefibrillar aggregates of both proteins, but not their mature fibrils or soluble monomers, impaired cholinergic neuron viability in a dose-dependent manner similar to that seen in cell cultures. Contrary to the situation with cultured cells, however, under our experimental conditions, cell stress in tissue is not followed by a comparable level of cell death, a result that is very likely to reflect the presence of protective mechanisms reducing aggregate toxicity. These findings support the hypothesis that neurodegenerative disorders result primarily from a generic cell dysfunction caused by early misfolded species in the aggregation process.


Subject(s)
Amyloid/toxicity , Brain/drug effects , Brain/pathology , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Proteins/toxicity , Animals , Carboxyl and Carbamoyl Transferases , Cells, Cultured , Dose-Response Relationship, Drug , Escherichia coli Proteins , Male , Neurodegenerative Diseases/chemically induced , Rats , Rats, Wistar , Toxins, Biological/toxicity
19.
Expert Rev Neurother ; 17(4): 345-358, 2017 04.
Article in English | MEDLINE | ID: mdl-27762153

ABSTRACT

INTRODUCTION: Clinical trials and population studies indicate the healthy virtues of the Mediterranean diet and its main lipid component, extra-virgin olive oil (EVOO). Olive leaves and EVOO contain many phenolics effective against several aging and lifestyle-related diseases, including neurodegeneration, both in animal models and in humans. Recent research has shown that such protection stems from several effects, including (i.) the interference with the aggregation of peptides/proteins found in amyloid diseases, particularly in Alzheimer's and Parkinson's diseases; (ii.) the protection of cells and tissues against aging-associated functional derangement (ion/redox homeostasis, aberrant cell signaling, etc.); (iii.) the transcriptional modulation through epigenetic modifications. Area covered: We used MEDLINE for literature reference; we also searched ClinicalTrials.gov to select clinical trials with olive oil and/or its polyphenols that suggested their potential particularly for what neuroprotective therapy is concerned. Expert commentary: We focus the relation between diet components, particularly olive polyphenols, and protection against the occurrence of the most widespread neurodegenerative conditions associated with aging. The need of more clinical studies in humans to confirm the results obtained in animal and cell models to definitely support the utility of these molecules to combat or to delay the symptoms associated to aging-associated neurodegeneration is also stressed.


Subject(s)
Aging , Brain Diseases , Olea , Polyphenols , Animals , Brain Diseases/drug therapy , Brain Diseases/physiopathology , Humans , Phenols , Plant Oils , Polyphenols/therapeutic use
20.
Front Cell Neurosci ; 11: 339, 2017.
Article in English | MEDLINE | ID: mdl-29163051

ABSTRACT

The degeneration of cholinergic neurons of the nucleus basalis of Meynert (NBM) in the basal forebrain (BF) is associated to the cognitive decline of Alzheimer's disease (AD) patients. To date no resolutive therapies exist. Cell-based replacement therapy is a strategy currently under consideration, although the mechanisms underlying the generation of stem cell-derived NBM cholinergic neurons able of functional integration remain to be clarified. Since fetal brain is an optimal source of neuronal cells committed towards a specific phenotype, this study is aimed at isolating cholinergic neurons from the human fetal NBM (hfNBMs) in order to study their phenotypic, maturational and functional properties. Extensive characterization confirmed the cholinergic identity of hfNBMs, including positivity for specific markers (such as choline acetyltransferase) and acetylcholine (Ach) release. Electrophysiological measurements provided the functional validation of hfNBM cells, which exhibited the activation of peculiar sodium (INa) and potassium (IK) currents, as well as the presence of functional cholinergic receptors. Accordingly, hfNBMs express both nicotinic and muscarinic receptors, which were activated by Ach. The hfNBMs cholinergic phenotype was regulated by the nerve growth factor (NGF), through the activation of the high-affinity NGF receptor TrkA, as well as by 17-ß-estradiol through a peculiar recruitment of its own receptors. When intravenously administered in NBM-lesioned rats, hfNBMs determined a significant improvement in memory functions. Histological examination of brain sections showed that hfNBMs (labeled with PKH26 fluorescent dye prior to administration) reached the damaged brain areas. The study provides a useful model to study the ontogenetic mechanisms regulating the development and maintenance of the human brain cholinergic system and to assess new lines of research, including disease modeling, drug discovery and cell-based therapy for AD.

SELECTION OF CITATIONS
SEARCH DETAIL