Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Microbes Infect ; : 105400, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39069117

ABSTRACT

Infection by SARS-CoV-2 is associated with uncontrolled inflammatory response during COVID-19 severe disease, in which monocytes are one of the main sources of pro-inflammatory mediators leading to acute respiratory distress syndrome. Extracellular vesicles (EVs) from different cells play important roles during SARS-CoV-2 infection, but investigations describing the involvement of EVs from primary human monocyte-derived macrophages (MDM) on the regulation of this infection are not available. Here, we describe the effects of EVs released by MDM stimulated with the neuropeptides VIP and PACAP on SARS-CoV-2-infected monocytes. MDM-derived EVs were isolated by differential centrifugation of medium collected from cells cultured for 24 h in serum-reduced conditions. Based on morphological properties, we distinguished two subpopulations of MDM-EVs, namely large (LEV) and small EVs (SEV). We found that MDM-derived EVs stimulated with the neuropeptides inhibited SARS-CoV-2 RNA synthesis/replication in monocytes, protected these cells from virus-induced cytopathic effects and reduced the production of pro-inflammatory mediators. In addition, EVs derived from VIP- and PACAP-treated MDM prevented the SARS-CoV-2-induced NF-κB activation. Overall, our findings suggest that MDM-EVs are endowed with immunoregulatory properties that might contribute to the antiviral and anti-inflammatory responses in SARS-CoV-2-infected monocytes and expand our knowledge of EV effects during COVID-19 pathogenesis.

2.
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
3.
Nat Med ; 25(1): 165-175, 2019 01.
Article in English | MEDLINE | ID: mdl-30617325

ABSTRACT

Defective brain hormonal signaling has been associated with Alzheimer's disease (AD), a disorder characterized by synapse and memory failure. Irisin is an exercise-induced myokine released on cleavage of the membrane-bound precursor protein fibronectin type III domain-containing protein 5 (FNDC5), also expressed in the hippocampus. Here we show that FNDC5/irisin levels are reduced in AD hippocampi and cerebrospinal fluid, and in experimental AD models. Knockdown of brain FNDC5/irisin impairs long-term potentiation and novel object recognition memory in mice. Conversely, boosting brain levels of FNDC5/irisin rescues synaptic plasticity and memory in AD mouse models. Peripheral overexpression of FNDC5/irisin rescues memory impairment, whereas blockade of either peripheral or brain FNDC5/irisin attenuates the neuroprotective actions of physical exercise on synaptic plasticity and memory in AD mice. By showing that FNDC5/irisin is an important mediator of the beneficial effects of exercise in AD models, our findings place FNDC5/irisin as a novel agent capable of opposing synapse failure and memory impairment in AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Fibronectins/metabolism , Memory Disorders/complications , Memory Disorders/physiopathology , Neuronal Plasticity , Physical Conditioning, Animal , Adolescent , Adult , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/genetics , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Down-Regulation , Female , Fibronectins/cerebrospinal fluid , Fibronectins/genetics , Humans , Long-Term Potentiation , Male , Mice, Inbred C57BL , Middle Aged , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction
4.
Eur J Pharmacol ; 586(1-3): 24-34, 2008 May 31.
Article in English | MEDLINE | ID: mdl-18371953

ABSTRACT

Gliomas are the most common and devastating tumors of the central nervous system. Several studies have suggested that nonsteroidal anti-inflammatory drugs (NSAIDs) are promising anticancer agents. Biodegradable nanoparticulate systems have received considerable attention as potential drug delivery vehicles. The aim of this study was to evaluate the effects of indomethacin-loaded nanocapsules and indomethacin ethyl ester-loaded nanocapsules on glioma cell lines. In addition, the effect of these formulations on normal neural tissue was also evaluated. In order to investigate this, glioma cell lines (U138-MG and C6) and hippocampal organotypic cultures were used. The main finding of the present study is that indomethacin-loaded nanocapsules formulation was more potent than a solution of indomethacin in decreasing the viability and cell proliferation of glioma lines. Indomethacin and indomethacin ethyl ester associated together in the same nanocapsule formulation caused a synergic effect decreasing glioma cell proliferation. In addition, when the glioma cells were exposed to 25 microM of indomethacin-loaded nanocapsules or indomethacin ethyl ester-loaded nanocapsules, a necrotic cell death was observed. Interestingly, 5 microM of indomethacin-loaded nanocapsules was able to cause an antiproliferative effect without promoting necrosis in glioma cells. Another important finding was that the cytotoxic effect induced by 25 microM or 50 microM of indomethacin-loaded nanocapsules or indomethacin ethyl ester-loaded nanocapsules, in glioma cells was not observed in the organotypic cultures, indicating selective cytotoxicity of those formulations for tumoral cells. Further investigations using in vivo glioma model should be helpful to confirm the distinct effects of indomethacin-loaded nanocapsules and indomethacin ethyl ester-loaded nanocapsules, in normal versus tumoral cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Antineoplastic Agents , Brain Neoplasms/drug therapy , Cyclooxygenase 2 Inhibitors/toxicity , Glioma/drug therapy , Indomethacin/analogs & derivatives , Indomethacin/toxicity , Animals , Brain Neoplasms/pathology , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Chemical Phenomena , Chemistry, Physical , Coloring Agents , Drug Compounding , Glioma/pathology , Hippocampus/drug effects , Humans , Hydrogen-Ion Concentration , Male , Nanocapsules , Organ Culture Techniques , Propidium , Rats , Rats, Wistar , Suspensions
5.
Front Neurosci ; 12: 37, 2018.
Article in English | MEDLINE | ID: mdl-29467605

ABSTRACT

Alzheimer's disease (AD), the most common form of dementia in late life, will become even more prevalent by midcentury, constituting a major global health concern with huge implications for individuals and society. Despite scientific breakthroughs during the past decades that have expanded our knowledge on the cellular and molecular bases of AD, therapies that effectively halt disease progression are still lacking, and focused efforts are needed to address this public health challenge. Because AD is classically recognized as a disease of memory, studies have mainly focused on investigating memory-associated brain defects. However, compelling evidence has indicated that additional brain regions, not classically linked to memory, are also affected in the course of disease. In this review, we outline the current understanding of key pathophysiological mechanisms in AD and their clinical manifestation. We also highlight how considering the complex nature of AD pathogenesis, and exploring repurposed drug approaches can pave the road toward the development of novel therapeutics for AD.

6.
J Alzheimers Dis ; 64(s1): S405-S426, 2018.
Article in English | MEDLINE | ID: mdl-29562518

ABSTRACT

Clinical trials have extensively failed to find effective treatments for Alzheimer's disease (AD) so far. Even after decades of AD research, there are still limited options for treating dementia. Mounting evidence has indicated that AD patients develop central and peripheral metabolic dysfunction, and the underpinnings of such events have recently begun to emerge. Basic and preclinical studies have unveiled key pathophysiological mechanisms that include aberrant brain stress signaling, inflammation, and impaired insulin sensitivity. These findings are in accordance with clinical and neuropathological data suggesting that AD patients undergo central and peripheral metabolic deregulation. Here, we review recent basic and clinical findings indicating that metabolic defects are central to AD pathophysiology. We further propose a view for future therapeutics that incorporates metabolic defects as a core feature of AD pathogenesis. This approach could improve disease understanding and therapy development through drug repurposing and/or identification of novel metabolic targets.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Animals , Humans
7.
Mol Neurobiol ; 55(1): 435-444, 2018 01.
Article in English | MEDLINE | ID: mdl-27966074

ABSTRACT

Sepsis survivors frequently develop late cognitive impairment. Because little is known on the mechanisms of post-septic memory deficits, there are no current effective approaches to prevent or treat such symptoms. Here, we subjected mice to severe sepsis induced by cecal ligation and puncture (CLP) and evaluated the sepsis-surviving animals in the open field, novel object recognition (NOR), and step-down inhibitory avoidance (IA) task at different times after surgery. Post-septic mice (30 days post-surgery) failed in the NOR and IA tests but exhibited normal performance when re-evaluated 45 days after surgery. Cognitive impairment in post-septic mice was accompanied by reduced hippocampal levels of proteins involved in synaptic plasticity, including synaptophysin, cAMP response element-binding protein (CREB), CREB phosphorylated at serine residue 133 (CREBpSer133), and GluA1 phosphorylated at serine residue 845 (GluA1pSer845). Expression of tumor necrosis factor α (TNF-α) was increased and brain insulin signaling was disrupted, as indicated by increased hippocampal IRS-1 phosphorylation at serine 636 (IRS-1pSer636) and decreased phosphorylation of IRS-1 at tyrosine 465 (IRS-1pTyr465), in the hippocampus 30 days after CLP. Phosphorylation of Akt at serine 473 (AktpSer473) and of GSK3 at serine 9 (GSK3ßpSer9) were also decreased in hippocampi of post-septic animals, further indicating that brain insulin signaling is disrupted by sepsis. We then treated post-septic mice with liraglutide, a GLP-1 receptor agonist with insulinotropic activity, or TDZD-8, a GSK3ß inhibitor, which rescued NOR memory. In conclusion, these results establish that hippocampal inflammation and disrupted insulin signaling are induced by sepsis and are linked to late memory impairment in sepsis survivors.


Subject(s)
Brain/metabolism , Cognitive Dysfunction/metabolism , Insulin/metabolism , Sepsis/metabolism , Signal Transduction/physiology , Animals , Brain/pathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Exploratory Behavior/physiology , Male , Mice , Sepsis/complications , Sepsis/pathology
8.
EMBO Mol Med ; 7(2): 190-210, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25617315

ABSTRACT

Alzheimer's disease (AD) is associated with peripheral metabolic disorders. Clinical/epidemiological data indicate increased risk of diabetes in AD patients. Here, we show that intracerebroventricular infusion of AD-associated Aß oligomers (AßOs) in mice triggered peripheral glucose intolerance, a phenomenon further verified in two transgenic mouse models of AD. Systemically injected AßOs failed to induce glucose intolerance, suggesting AßOs target brain regions involved in peripheral metabolic control. Accordingly, we show that AßOs affected hypothalamic neurons in culture, inducing eukaryotic translation initiation factor 2α phosphorylation (eIF2α-P). AßOs further induced eIF2α-P and activated pro-inflammatory IKKß/NF-κB signaling in the hypothalamus of mice and macaques. AßOs failed to trigger peripheral glucose intolerance in tumor necrosis factor-α (TNF-α) receptor 1 knockout mice. Pharmacological inhibition of brain inflammation and endoplasmic reticulum stress prevented glucose intolerance in mice, indicating that AßOs act via a central route to affect peripheral glucose homeostasis. While the hypothalamus has been largely ignored in the AD field, our findings indicate that AßOs affect this brain region and reveal novel shared molecular mechanisms between hypothalamic dysfunction in metabolic disorders and AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Hypothalamus/metabolism , Oligonucleotides/metabolism , Peripheral Nerves/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Female , Glucose/metabolism , Humans , Macaca , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Neurons/metabolism , Oligonucleotides/genetics , Rats , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
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
10.
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
11.
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
12.
J Biomed Nanotechnol ; 9(3): 516-26, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23621009

ABSTRACT

The development of novel therapeutic strategies to treat gliomas remains critical as a result of the poor prognoses, inef-. ficient therapies and recurrence associated with these tumors. In this context, biodegradable nanoparticles are emerging as efficient drug delivery systems for the treatment of difficult-to-treat diseases such as brain tumors. In the current study, we evaluated the antiglioma effect of trans-resveratrol-loaded lipid-core nanocapsules (RSV-LNC) based on in vitro (C6 glioma cell line) and in vivo (brain-implanted C6 cells) models of the disease. In vitro, RSV-LNC decreased the viability of C6 glioma cells to a higher extent than resveratrol in solution. Interestingly, RSV-LNC treatment was not cytotoxic to hippocampal organotypic cultures, a model of healthy neural cells, suggesting selectivity for cancer cells. RSV-LNC induced losses in glioma cell viability through induction of apoptotic cell death, as assessed by Annexin-FITC/PI assay, which was preceded by an early arrest in the S and G1 phases of the cell cycle. In brain-implanted C6 tumors, treatment with RSV-LNC (5 mg/kg/day, i.p.) for 10 days promoted a marked decrease in tumor size and also reduced the incidence of some malignant tumor-associated characteristics, such as intratumoral hemorrhaging, intratumoral edema and pseudopalisading, compared to resveratrol in solution. Taken together, the results presented herein suggest that nanoencapsulation of resveratrol improves its antiglioma activity, thus providing a provocative foundation for testing the clinical usefulness of nanoformulations of this natural compound as a new chemotherapeutic strategy for the treatment of gliomas.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Glioma/drug therapy , Glioma/pathology , Lipids/chemistry , Nanocapsules/chemistry , Stilbenes/therapeutic use , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemical Phenomena , Chemistry, Pharmaceutical , Disease Models, Animal , G1 Phase/drug effects , Hippocampus/drug effects , Humans , Male , Neoplasm Transplantation , Rats , Rats, Wistar , Resveratrol , S Phase/drug effects , Solutions , Stilbenes/pharmacology , Tumor Burden/drug effects
13.
Cell Metab ; 18(6): 831-43, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24315369

ABSTRACT

Alzheimer's disease (AD) and type 2 diabetes appear to share similar pathogenic mechanisms. dsRNA-dependent protein kinase (PKR) underlies peripheral insulin resistance in metabolic disorders. PKR phosphorylates eukaryotic translation initiation factor 2α (eIF2α-P), and AD brains exhibit elevated phospho-PKR and eIF2α-P levels. Whether and how PKR and eIF2α-P participate in defective brain insulin signaling and cognitive impairment in AD are unknown. We report that ß-amyloid oligomers, AD-associated toxins, activate PKR in a tumor necrosis factor α (TNF-α)-dependent manner, resulting in eIF2α-P, neuronal insulin receptor substrate (IRS-1) inhibition, synapse loss, and memory impairment. Brain phospho-PKR and eIF2α-P were elevated in AD animal models, including monkeys given intracerebroventricular oligomer infusions. Oligomers failed to trigger eIF2α-P and cognitive impairment in PKR(-/-) and TNFR1(-/-) mice. Bolstering insulin signaling rescued phospho-PKR and eIF2α-P. Results reveal pathogenic mechanisms shared by AD and diabetes and establish that proinflammatory signaling mediates oligomer-induced IRS-1 inhibition and PKR-dependent synapse and memory loss.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/drug effects , Insulin Receptor Substrate Proteins/metabolism , Polymers/toxicity , Tumor Necrosis Factor-alpha/metabolism , eIF-2 Kinase/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Animals , Brain/metabolism , Disease Models, Animal , Haplorhini/metabolism , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/antagonists & inhibitors , Memory Disorders/metabolism , Memory Disorders/pathology , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Polymers/chemistry , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Synapses/drug effects , Synapses/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , eIF-2 Kinase/deficiency , eIF-2 Kinase/genetics
14.
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
15.
Neurochem Int ; 59(5): 648-55, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21723896

ABSTRACT

The effect of Aß25-35 peptide, in its fibrillar and non-fibrillar forms, on ganglioside expression in organotypic hippocampal slice cultures was investigated. Gangliosides were endogenously labeled with D-[1-C(14)] galactose and results showed that Aß25-35 affected ganglioside expression, depending on the peptide aggregation state, that is, fibrillar Aß25-35 caused an increase in GM3 labeling and a reduction in GD1b labeling, whereas the non-fibrillar form was able to enhance GM1 expression. Interestingly, GM1 exhibited a neuroprotective effect in this organotypic model, since pre-treatment of the hippocampal slices with GM1 10 µM was able to prevent the toxicity triggered by the fibrillar Aß25-35, when measured by propidium iodide uptake protocol. With the purpose of further investigating a possible mechanism of action, we analyzed the effect of GM1 treatment (1, 6, 12 and 24h) upon the Aß-induced alterations on GSK3ß dephosphorylation/activation state. Results demonstrated an important effect after 24-h incubation, with GM1 preventing the Aß-induced dephosphorylation (activation) of GSK3ß, a signaling pathway involved in apoptosis triggering and neuronal death in models of Alzheimer's disease. Taken together, present results provide a new and important support for ganglioside participation in development of Alzheimer's disease experimental models and suggest a protective role for GM1 in Aß-induced toxicity. This may be useful for designing new therapeutic strategies for Alzheimer's treatment.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/toxicity , G(M1) Ganglioside/pharmacology , Gangliosides/biosynthesis , Neuroprotective Agents , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/toxicity , Animals , Blotting, Western , Cell Death/drug effects , Chromatography, Thin Layer , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/metabolism , Lipid Metabolism/drug effects , Male , Neurofibrils/drug effects , Neurofibrils/metabolism , Organ Culture Techniques , Phosphorylation , Rats , Rats, Wistar
16.
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
17.
Neurochem Int ; 57(6): 629-36, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20691236

ABSTRACT

Targeted treatment of diseases of the central nervous system remains problematic due to the complex pathogenesis of these disorders and the difficulty in drug delivery. Here we investigate the neuroprotective effect of indomethacin-loaded nanocapsules (IndOH-NC) in an in vitro model of ischemia. For this purpose we used organotypic hippocampal cultures exposed to oxygen-glucose deprivation (OGD). When the cultures were exposed to 60 min of OGD, 54.5±14.7% of the total area of the hippocampal slices was labeled with propidium iodide. On the other hand, when the cultures were treated with 50 or 100 µM of IndOH-NC the cell death was significantly reduced to 31±7% (P<0.05) and 20±4% (P<0.001), respectively. The treatment with IndOH-NC markedly inhibited the levels of the pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α levels even 48 h after OGD. Immunoblotting revealed that treatment with 100 µM of IndOH-NC was able to significantly reduce to the levels of control cultures the levels of ERK1/2 and JNK phosphorylation, as well as iNOS activation. Additionally, IndOH-NC prevented glial activation induced by OGD, as evidenced by a decrease of GFAP immunocontent and Isolectin B(4) reactivity. Our results clearly demonstrate that IndOH-NC might represent a promising pharmaceutical neuroprotective formulation for cerebral ischemia, most probably by inhibiting the inflammatory cascades.


Subject(s)
Glucose/metabolism , Hippocampus/drug effects , Indomethacin/pharmacology , Inflammation/metabolism , Nanocapsules , Neuroprotective Agents/pharmacology , Animals , Blotting, Western , Cytokines/metabolism , Enzyme Activation , Enzyme Induction , Hippocampus/enzymology , Hippocampus/metabolism , In Vitro Techniques , Inflammation Mediators/metabolism , Protein Kinases/biosynthesis , Protein Kinases/metabolism
18.
Neurobiol Dis ; 24(1): 170-82, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16860989

ABSTRACT

Here we investigated the neuroprotective effect of resveratrol in an in vitro model of ischemia. We used organotypic hippocampal cultures exposed to oxygen-glucose deprivation (OGD). In OGD-vehicle exposed cultures, about 46% of the hippocampus was labeled with PI, indicating a robust percentage of cell death. When cultures were treated with resveratrol 10, 25 and 50 microM, the cell death was reduced to 22, 20 and 13% respectively. To elucidate a possible mechanism by which resveratrol exerts its neuroprotective effect, we investigated the phosphoinositide3-kinase (PI3-k) pathway using LY294002 (5 microM) and mitogen-activated protein kinase (MAPK) using PD98059 (20 microM). The resveratrol (50 microM) neuroprotection was prevented by LY294002 but was not by PD98059. Immunoblotting revealed that resveratrol 50 microM induced the phosphorylation/activation of Akt and extracellular signal-regulated kinase-1 and -2 (ERK1/2) and the phosphorylation/inactivation of glycogen synthase kinase-3beta (GSK-3beta). Our results suggest that PI3-k/Akt pathway are involved in the neuroprotective effect of resveratrol.


Subject(s)
Glucose/deficiency , Hippocampus/physiology , Hypoxia, Brain/pathology , Neuroprotective Agents , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology , Stilbenes/pharmacology , Animals , Animals, Newborn , Blotting, Western , Cell Count , Cell Death/drug effects , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Male , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/physiology , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/physiology , Organ Culture Techniques , Phosphoinositide-3 Kinase Inhibitors , Rats , Rats, Wistar , Resveratrol , Signal Transduction/drug effects , Stilbenes/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL