Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Aging Dis ; 6(5): 322-30, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26425387

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized in the brain by the formation of amyloid-beta (Aß)-containing plaques and neurofibrillary tangles containing the microtubule-associated protein tau. Neuroinflammation is another feature of AD and astrocytes are receiving increasing attention as key contributors. Although some progress has been made, the molecular mechanisms underlying the pathophysiology of AD remain unclear. Interestingly, some of the main proteins involved in AD, including amyloid precursor protein (APP) and tau, have recently been shown to be SUMOylated. The post-translational modification by SUMO (small ubiquitin-like modifier) has been shown to regulate APP and tau and may modulate other proteins implicated in AD. Here we present an overview of recent studies suggesting that protein SUMOylation might be involved in the underlying pathogenic mechanisms of AD and discuss how this could be exploited for therapeutic intervention.

2.
Eur J Med Chem ; 95: 552-62, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25863023

ABSTRACT

We described the first synthesis of fatty acid 3,4-dihydropyrimidinones (DHPM-fatty acids) using the Biginelli multicomponent reaction. Antiproliferative activity on two glioma cell lines (C6 rat and U-138-MG human) was also reported. The novel DHPM-fatty acids reduced glioma cell viability relative to temozolomide. Hybrid oxo-monastrol-palmitic acid was the most potent, reducing U-138-MG human cell viability by ca. 50% at 10 µM. In addition, the DHPM-fatty acids showed a large safety range to neural cells, represented by the organotypic hippocampal culture. These results suggest that the increased lipophilicity of DHPM-fatty acids offer a promising approach to overcoming resistance to chemotherapy and may play an important role in the development of new antitumor drugs.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Fatty Acids/chemical synthesis , Fatty Acids/pharmacology , Glioma/pathology , Uridine/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Drug Design , Fatty Acids/chemistry , Humans , Male , Rats , Rats, Wistar , Uridine/chemistry
3.
J Biomed Nanotechnol ; 9(12): 2086-104, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24266263

ABSTRACT

Resveratrol, a natural polyphenolic compound, has attracted considerable interest for its anti-inflammatory and neuroprotective properties. However, the biological effects of resveratrol appear strongly limited because it is photosensitive, easily oxidized, and has unfavorable pharmacokinetics. The present study aimed to elucidate the effect of resveratrol on Abeta-triggered neuroinflammation by comparing the effects of free resveratrol (RSV) treatment with those of treatment with resveratrol-loaded lipid-core nanocapsules (RSV-LNC). Organotypic hippocampal cultures were stimulated by Abeta1-42 with or without different concentrations of RSV or RSV-LNC. We found that Abeta triggered a harmful neuroinflammation process in organotypic hippocampal cultures. Pre- and co-treatments with RSV-LNC were able to protect cultures against ROS formation and cell death induced by Abeta, possibly through sustained blocking of TNF-alpha, IL-1beta, and IL-6 release. Furthermore, RSV-LNC was able to increase IL-10 release even in the presence of Abeta and prevent or decrease both glial and JNK activation. On the other hand, both pre- and co-treatment with RSV exhibited a lower ability to prevent or decrease neuroinflammation, ROS formation, and cell death, and failed to increase IL-10 release. Our findings suggest that modulation of neuroinflammation through a combination of resveratrol and a lipid-core nanocapsule-based delivery system might represent a promising approach for preventing or delaying the neurodegenerative process triggered by Abeta. The results open new vistas to the interplay between inflammation and amyloid pathology.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/prevention & control , Lipids/pharmacology , Nanocapsules/chemistry , Neurons/drug effects , Stilbenes/pharmacology , Amyloid beta-Peptides , Animals , Anti-Inflammatory Agents/administration & dosage , Cells, Cultured , Drug Synergism , Encephalitis/chemically induced , Encephalitis/pathology , Encephalitis/prevention & control , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipids/administration & dosage , Lipids/chemistry , Male , Neurons/pathology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Resveratrol , Stilbenes/administration & dosage
4.
Neurobiol Learn Mem ; 106: 134-44, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23954730

ABSTRACT

Alzheimer's disease (AD), a neurodegenerative disorder exhibiting progressive loss of memory and cognitive functions, is characterized by the presence of neuritic plaques composed of neurofibrillary tangles and ß-amyloid (Aß) peptide. Drug delivery to the brain still remains highly challenging for the treatment of AD. Several studies have been shown that curcumin is associated with anti-amyloidogenic properties, but therapeutic application of its beneficial effects is limited. Here we investigated possible mechanisms involved in curcumin protection against Aß(1-42)-induced cognitive impairment and, due to its poor bioavailability, we developed curcumin-loaded lipid-core nanocapsules in an attempt to improve the neuroprotective effect of this polyphenol. Animals received a single intracerebroventricular injection of Aß(1-42) and they were administered either free curcumin or curcumin-loaded lipid-core nanocapsules (Cur-LNC) intraperitoneally for 10days. Aß(1-42)-infused animals showed a significant impairment on learning-memory ability, which was paralleled by a significant decrease in hippocampal synaptophysin levels. Furthermore, animals exhibited activated astrocytes and microglial cells, as well as disturbance in BDNF expression and Akt/GSK-3ß signaling pathway, beyond tau hyperphosphorylation. Our findings demonstrate that administration of curcumin was effective in preventing behavioral impairments, neuroinflammation, tau hyperphosphorylation as well as cell signaling disturbances triggered by Aß in vivo. Of high interest, Cur-LNC in a dose 20-fold lower presented similar neuroprotective results compared to the effective dose of free curcumin. Considered overall, the data suggest that curcumin is a potential therapeutic agent for neurocognition and nanoencapsulation of curcumin in LNC might constitute a promising therapeutic alternative in the treatment of neurodegenerative diseases such as AD.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/drug therapy , Curcumin/therapeutic use , Glycogen Synthase Kinase 3/metabolism , Memory/drug effects , Neuroprotective Agents/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Amyloid beta-Peptides , Animals , Cognition/drug effects , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Curcumin/pharmacology , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Synaptophysin/metabolism
5.
Neurosci Lett ; 546: 51-6, 2013 Jun 24.
Article in English | MEDLINE | ID: mdl-23651519

ABSTRACT

Astrocyte reactivity is implicated in the neuronal loss underlying Alzheimer's disease. Curcumin has been shown to reduce astrocyte reactivity, though the exact pathways underlying these effects are incompletely understood. Here we investigated the role of the small ubiquitin-like modifier (SUMO) conjugation in mediating this effect of curcumin. In beta-amyloid (Aß)-treated astrocytes, morphological changes and increased glial fibrillary acidic protein (GFAP) confirmed reactivity, which was accompanied by c-jun N-terminal kinase activation. Moreover, the levels of SUMO-1 conjugated proteins, as well as the conjugating enzyme, Ubc9, were decreased, with concomitant treatment with curcumin preventing these effects. Increasing SUMOylation in astrocytes, by over-expression of constitutively active SUMO-1, but not its inactive mutant, abrogated Aß-induced increase in GFAP, suggesting astrocytes require SUMO-1 conjugation to remain non-reactive.


Subject(s)
Amyloid beta-Peptides/pharmacology , Astrocytes/physiology , Curcumin/pharmacology , MAP Kinase Kinase 4/metabolism , SUMO-1 Protein/metabolism , Sumoylation/physiology , Animals , Astrocytes/drug effects , Cells, Cultured , Drug Interactions , Enzyme Activation/drug effects , Enzyme Activation/physiology , Mice , Sumoylation/drug effects
6.
Int J Nanomedicine ; 8: 711-28, 2013.
Article in English | MEDLINE | ID: mdl-23440594

ABSTRACT

Despite recent advances in radiotherapy, chemotherapy, and surgical techniques, glioblastoma multiforme (GBM) prognosis remains dismal. There is an urgent need for new therapeutic strategies. Nanoparticles of biodegradable polymers for anticancer drug delivery have attracted intense interest in recent years because they can provide sustained, controlled, and targeted delivery. Here, we investigate the mechanisms involved in the antiproliferative effect of indomethacin-loaded lipid-core nanocapsules (IndOH-LNC) in glioma cells. IndOH-LNC were able to reduce cell viability by inducing apoptotic cell death in C6 and U138-MG glioma cell lines. Interestingly, IndOH-LNC did not affect the viability of primary astrocytes, suggesting that this formulation selectively targeted transformed cells. Mechanistically, IndOH-LNC induced inhibition of cell growth and cell-cycle arrest to be correlated with the inactivation of AKT and ß-catenin and the activation of GSK-3ß. IndOH-LNC also induced G0/G1 and/or G2/M phase arrest, which was accompanied by a decrease in the levels of cyclin D1, cyclin B1, pRb, and pcdc2 and an increase in the levels of Wee1 CDK inhibitor p21(WAF1). Additionally, IndOH-LNC promoted GBM cell differentiation, observed as upregulation of glial fibrillary acidic protein (GFAP) protein and downregulation of nestin and CD133. Taken together, the crosstalk among antiproliferative effects, cell-cycle arrest, apoptosis, and cell differentiation should be considered when tailoring pharmacological interventions aimed at reducing glioma growth by using formulations with multiples targets, such as IndOH-LNC.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Indomethacin/administration & dosage , Nanocapsules/administration & dosage , Analysis of Variance , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Indomethacin/chemistry , Nanocapsules/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , beta Catenin/metabolism
7.
Mol Neurobiol ; 47(3): 1066-80, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23315270

ABSTRACT

Alzheimer's disease (AD), a neurodegenerative disorder exhibiting a gradual decline in cognitive function, is characterized by the presence of neuritic plaques composed of neurofibrillary tangles and amyloid-ß (Aß) peptide. Available drugs for AD therapy have small effect sizes and do not alter disease progression. Several studies have been shown that resveratrol is associated with anti-amyloidogenic properties, but therapeutic application of its beneficial effects is limited. Here we compared the neuroprotective effects of free resveratrol treatment with those of resveratrol-loaded lipid-core nanocapsule treatment against intracerebroventricular injection of Aß1-42 in rats. Animals received a single intracerebroventricular injection of Aß1-42 (2 nmol), and 1 day after Aß infusion, they were administered either free resveratrol (RSV) or resveratrol-loaded lipid-core nanocapsules (5 mg/kg, each 12 h, intraperitoneally), for 14 days. Aß1-42-infused animals showed a significant impairment on learning memory ability, which was paralleled by a significant decrease in hippocampal synaptophysin levels. Furthermore, animals exhibited activated astrocytes and microglial cells, as well as disturbance in c-Jun N-terminal kinase (JNK) and glycogen synthase kinase-3ß (GSK-3ß) activation, beyond destabilization of ß-catenin levels. Our results clearly show that by using lipid-core nanocapsules, resveratrol was able to rescue the deleterious effects of Aß1-42 while treatment with RSV presented only partial beneficial effects. These findings might be explained by the robust increase of resveratrol concentration in the brain tissue achieved by lipid-core nanocapsules. Our data not only confirm the potential of resveratrol in treating AD but also offer an effective way to improve the efficiency of resveratrol through the use of nanodrug delivery systems.


Subject(s)
Amyloid beta-Peptides/administration & dosage , Lipids/chemistry , Nanocapsules/chemistry , Neuroprotective Agents/pharmacology , Stilbenes/pharmacology , Amyloid beta-Peptides/toxicity , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Cytoprotection/drug effects , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/pathology , Injections, Intraventricular , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Memory Disorders/drug therapy , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neuroprotective Agents/therapeutic use , Protein Stability/drug effects , Rats , Rats, Wistar , Resveratrol , Signal Transduction/drug effects , Stilbenes/adverse effects , Stilbenes/therapeutic use , Synapses/drug effects , Synapses/pathology , Tissue Distribution/drug effects , beta Catenin/metabolism
8.
Int J Nanomedicine ; 7: 4927-42, 2012.
Article in English | MEDLINE | ID: mdl-23028221

ABSTRACT

Neuroinflammation, characterized by the accumulation of activated microglia and reactive astrocytes, is believed to modulate the development and/or progression of Alzheimer's disease (AD). Epidemiological studies suggesting that nonsteroidal anti-inflammatory drugs decrease the risk of developing AD have encouraged further studies elucidating the role of inflammation in AD. Nanoparticles have become an important focus of neurotherapeutic research because they are an especially effective form of drug delivery. Here, we investigate the potential protective effect of indomethacin-loaded lipid-core nanocapsules (IndOH-LNCs) against cell damage and neuroinflammation induced by amyloid beta (Aß)1-42 in AD models. Our results show that IndOH-LNCs attenuated Aß-induced cell death and were able to block the neuroinflammation triggered by Aß1-42 in organotypic hippocampal cultures. Additionally, IndOH-LNC treatment was able to increase interleukin-10 release and decrease glial activation and c-jun N-terminal kinase phosphorylation. As a model of Aß-induced neurotoxicity in vivo, animals received a single intracerebroventricular injection of Aß1-42 (1 nmol/site), and 1 day after Aß1-42 infusion, they were administered either free IndOH or IndOH-LNCs (1 mg/kg, intraperitoneally) for 14 days. Only the treatment with IndOH-LNCs significantly attenuated the impairment of this behavior triggered by intracerebroventricular injection of Aß1-42. Further, treatment with IndOH-LNCs was able to block the decreased synaptophysin levels induced by Aß1-42 and suppress glial and microglial activation. These findings might be explained by the increase of IndOH concentration in brain tissue attained using drug-loaded lipid-core NCs. All these findings support the idea that blockage of neuroinflammation triggered by Aß is involved in the neuroprotective effects of IndOH-LNCs. These data provide strong evidence that IndOH-LNC treatment may represent a promising approach for treating AD.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides , Disease Models, Animal , Indomethacin/administration & dosage , Lipids/chemistry , Nanocapsules/administration & dosage , Peptide Fragments , Alzheimer Disease/chemically induced , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Humans , Indomethacin/chemistry , Male , Nanocapsules/chemistry , Rats, Wistar , Treatment Outcome
9.
Int J Dev Neurosci ; 30(5): 369-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22525229

ABSTRACT

Homocysteine is a neurotoxic amino acid that accumulates in several disorders including homocystinuria, neurodegenerative and neuroinflammatory diseases. In the present study we evaluated the effect of acute and chronic hyperhomocysteinemia on Akt, NF-κB/p65, GSK-3ß, as well as Tau protein in hippocampus of rats. For acute treatment, rats received a single injection of homocysteine (0.6 µmol/g body weight) or saline (control). For chronic treatment, rats received daily subcutaneous injections of homocysteine (0.3-0.6 µmol/g body weight) or saline (control) from the 6th to the 28th days-of-age. One or 12h after the last injection, rats were euthanized, the hippocampus was removed and samples were submitted to electrophoresis followed by Western blotting. Results showed that acute hyperhomocysteinemia increases Akt phosphorylation, cytosolic and nuclear immunocontent of NF-κB/p65 subunit and Tau protein phosphorylation, but reduces GSK-3ß phosphorylation at 1h after homocysteine injection. However, 12h after acute hyperhomocysteinemia there is no effect on Akt and GSK-3ß phosphorylation. Furthermore, chronic hyperhomocysteinemia did not alter Akt and GSK-3ß phosphorylation at 1h and 12h after the last administration of this amino acid. Our data showed that Akt, NF-κB/p65, GSK-3ß and Tau protein are activated in hippocampus of rats subjected to acute hyperhomocysteinemia, suggesting that these signaling pathways may be, at least in part, important contributors to the neuroinflammation and/or brain dysfunction observed in some hyperhomocystinuric patients.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Glycogen Synthase Kinase 3/metabolism , Hyperhomocysteinemia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Disease Models, Animal , Gene Expression Regulation, Enzymologic/drug effects , Glycogen Synthase Kinase 3 beta , Homocysteine/adverse effects , Hyperhomocysteinemia/chemically induced , NF-kappa B/metabolism , Phosphorylation/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects , Time Factors , tau Proteins/metabolism
10.
J Biomed Nanotechnol ; 6(6): 694-703, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21361135

ABSTRACT

Several studies have reported that orally ingested trans-resveratrol is extensively metabolized in the enterocyte before it enters the blood and target organs. Additionally, trans-resveratrol is photosensitive, easily oxidized and presents unfavorable pharmacokinetics. Therefore, it is of great interest to stabilize trans-resveratrol in order to preserve its biological activities and to improve its bioavailability in the brain. Here, trans-resveratrol was loaded into lipid-core nanocapsules and analyzed for particle size, polydispersity and zeta potential. The nanocapsule distribution in brain tissue was evaluated by intraperitoneal (i.p.) and gavage routes in healthy rats. The lipid-core nanocapsules had a mean diameter of 241 nm, a polydispersity index of 0.2, and a zeta potential of -15 mV. No physical changes were observed after 1, 2 and 3 months of storage at 25 degrees C. Lipid-core nanocapsules showed high entrapment of trans-resveratrol and displayed a higher trans-resveratrol concentration in the brain, the liver and the kidney after daily i.p. or gavage administration than that observed for the free trans-resveratrol. Because trans-resveratrol is a potent cyclooxygenase-1 inhibitor, gastrointestinal damage was evaluated. The animals that were administered with trans-resveratrol-loaded lipid-core nanocapsules showed significantly less damage when compared to those administered with free trans-resveratrol. In summary, lipid-core nanocapsules exhibited great trans-resveratrol encapsulation efficiency. trans-Resveratrol-loaded lipid-core nanocapsules increased the concentration of trans-resveratrol in the brain tissue. Gastrointestinal safety was improved when compared with free trans-resveratrol. Thus, trans-resveratrol-loaded lipid-core nanocapsules may be used as an alternative potential therapeutic for several diseases including Alzheimer's disease.


Subject(s)
Antioxidants/pharmacokinetics , Lipids , Nanocapsules , Stilbenes/pharmacokinetics , Animals , Antioxidants/pharmacology , Drug Stability , Gastrointestinal Tract/drug effects , Hydrogen-Ion Concentration , Lipids/chemistry , Male , Nanocapsules/chemistry , Rats , Rats, Wistar , Resveratrol , Stilbenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...