Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Integr Neurosci ; 22(3): 75, 2023 May 16.
Article in English | MEDLINE | ID: mdl-37258443

ABSTRACT

BACKGROUND: Epilepsy is one of the most common neurologic diseases, and around 30% of all epilepsies, particularly the temporal lobe epilepsy (TLE), are highly refractory to current pharmacological treatments. Abnormal synchronic neuronal activity, brain glucose metabolism alterations, neurodegeneration and neuroinflammation are features of epilepsy. Further, neuroinflammation has been shown to contribute to dysregulation of neuronal excitability and the progression of epileptogenesis. Flufenamic acid (FLU), a non-steroidal anti-inflammatory drug, is also characterized by its wide properties as a dose-dependent ion channel modulator. In this context, in vitro studies have shown that it abolishes seizure-like events in neocortical slices stimulated with a gamma-aminobutyric acid A (GABAA) receptor blocker. However, little is known about its effects in animal models. Thus, our goal was to assess the efficacy and safety of a relatively high dose of FLU in the lithium-pilocarpine rat model of status epilepticus (SE). This animal model reproduces many behavioral and neurobiological features of TLE such as short-term brain hypometabolism, severe hippocampal neurodegeneration and inflammation reflected by a marked reactive astrogliosis. METHODS: FLU (100 mg/kg, i.p.) was administered to adult male rats, 150 min before SE induced by pilocarpine. Three days after the SE, brain glucose metabolism was assessed by 2-deoxy-2-[18F]-fluoro-D-glucose ([18F]FDG) positron emission tomography (PET). Markers of hippocampal integrity, neurodegeneration and reactive astrogliosis were also evaluated. RESULTS: FLU neither prevented the occurrence of the SE nor affected brain glucose hypometabolism as assessed by [18F]FDG PET. Regarding the neurohistochemical studies, FLU neither prevented neuronal damage nor hippocampal reactive astrogliosis. On the contrary, FLU increased the mortality rate and negatively affected body weight in the rats that survived the SE. CONCLUSIONS: Our results do not support an acute anticonvulsant effect of a single dose of FLU. Besides, FLU did not show short-term neuroprotective or anti-inflammatory effects in the rat lithium-pilocarpine model of SE. Moreover, at the dose administered, FLU resulted in deleterious effects.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Status Epilepticus , Rats , Male , Animals , Lithium/adverse effects , Pilocarpine/adverse effects , Flufenamic Acid/metabolism , Flufenamic Acid/pharmacology , Flufenamic Acid/therapeutic use , Rats, Sprague-Dawley , Fluorodeoxyglucose F18/metabolism , Fluorodeoxyglucose F18/pharmacology , Fluorodeoxyglucose F18/therapeutic use , Gliosis/metabolism , Neuroinflammatory Diseases , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/metabolism , Epilepsy/metabolism , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/diagnostic imaging , Epilepsy, Temporal Lobe/drug therapy , Hippocampus/metabolism , Glucose/metabolism , Anti-Inflammatory Agents/adverse effects , Disease Models, Animal
2.
Eur J Pharmacol ; 939: 175453, 2023 Jan 15.
Article in English | MEDLINE | ID: mdl-36516936

ABSTRACT

Status epilepticus (SE) triggered by lithium-pilocarpine is a model of epileptogenesis widely used in rats, reproducing many of the pathological features of human temporal lobe epilepsy (TLE). After the SE, a silent period takes place that precedes the occurrence of recurrent spontaneous seizures. This latent stage is characterized by brain glucose hypometabolism and intense neuronal damage, especially at the hippocampus. Importantly, interictal hypometabolism in humans is a predictive marker of epileptogenesis, being correlated to the extent and severity of neuronal damage. Among the potential mechanisms underpinning glucose metabolism impairment and the subsequent brain damage, a reduction of cerebral blood flow has been proposed. Accordingly, our goal was to evaluate the potential beneficial effects of naftidrofuryl (25 mg/kg i.p., twice after the insult), a vasodilator drug currently used for circulatory insufficiency-related pathologies. Thus, we measured the effects of naftidrofuryl on the short-term brain hypometabolism and hippocampal damage induced by SE in rats. 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET) neuroimaging along with various neurohistochemical assays aimed to assess brain damage were performed. SE led to both severe glucose hypometabolism in key epilepsy-related areas and hippocampal neuronal damage. Although naftidrofuryl showed no anticonvulsant properties, it ameliorated the short-term brain hypometabolism induced by pilocarpine. Strikingly, the latter was neither accompanied by neuroprotective nor by anti-inflammatory effects. We suggest that naftidrofuryl, by acutely enhancing brain blood flow around the time of SE improves the brain metabolic state but this effect is not enough to protect from the damage induced by SE.


Subject(s)
Nafronyl , Status Epilepticus , Humans , Rats , Animals , Pilocarpine/pharmacology , Lithium/pharmacology , Nafronyl/metabolism , Nafronyl/pharmacology , Vasodilator Agents/pharmacology , Neuroprotection , Glucose/metabolism , Disease Models, Animal , Brain , Status Epilepticus/chemically induced , Status Epilepticus/diagnostic imaging , Status Epilepticus/drug therapy , Hippocampus , Seizures/metabolism
3.
Planta Med ; 89(4): 364-376, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36130709

ABSTRACT

Numerous preclinical studies provide evidence that curcumin, a polyphenolic phytochemical extracted from Curcuma longa (turmeric) has neuroprotective, anti-inflammatory and antioxidant properties against various neurological disorders. Curcumin neuroprotective effects have been reported in different animal models of epilepsy, but its potential effect attenuating brain glucose hypometabolism, considered as an early marker of epileptogenesis that occurs during the silent period following status epilepticus (SE), still has not been addressed. To this end, we used the lithium-pilocarpine rat model to induce SE. Curcumin was administered orally (300 mg/kg/day, for 17 days). Brain glucose metabolism was evaluated in vivo by 2-deoxy-2-[18F]Fluoro-D-Glucose ([18F]FDG) positron emission tomography (PET). In addition, hippocampal integrity, neurodegeneration, microglia-mediated neuroinflammation, and reactive astrogliosis were evaluated as markers of brain damage. SE resulted in brain glucose hypometabolism accompanied by body weight (BW) loss, hippocampal neuronal damage, and neuroinflammation. Curcumin did not reduce the latency time to the SE onset, nor the mortality rate associated with SE. Nevertheless, it reduced the number of seizures, and in the surviving rats, curcumin protected BW and attenuated the short-term glucose brain hypometabolism as well as the signs of neuronal damage and neuroinflammation induced by the SE. Overall, our results support the potential adaptogen-like effects of curcumin attenuating key features of SE-induced brain damage.


Subject(s)
Curcumin , Status Epilepticus , Rats , Animals , Curcumin/pharmacology , Curcumin/metabolism , Rats, Sprague-Dawley , Neuroinflammatory Diseases , Brain , Hippocampus , Status Epilepticus/chemically induced , Status Epilepticus/diagnostic imaging , Status Epilepticus/drug therapy , Positron-Emission Tomography/methods , Glucose/pharmacology , Pilocarpine/metabolism , Pilocarpine/pharmacology , Disease Models, Animal
4.
Pharmaceutics ; 14(11)2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36365169

ABSTRACT

Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, with its incidence constantly increasing. To date, there is no cure for the disease, with a need for new and effective treatments. Morin hydrate (MH) is a naturally occurring flavonoid of the Moraceae family with antioxidant and anti-inflammatory properties; however, the blood-brain barrier (BBB) prevents this flavonoid from reaching the CNS when aiming to potentially treat AD. Seeking to use the LAT-1 transporter present in the BBB, a nanoparticle (NPs) formulation loaded with MH and functionalized with phenylalanine-phenylalanine dipeptide was developed (NPphe-MH) and compared to non-functionalized NPs (NP-MH). In addition, two formulations were prepared using rhodamine B (Rh-B) as a fluorescent dye (NPphe-Rh and NP-Rh) to study their biodistribution and ability to cross the BBB. Functionalization of PLGA NPs resulted in high encapsulation efficiencies for both MH and Rh-B. Studies conducted in Wistar rats showed that the presence of phenylalanine dipeptide in the NPs modified their biodistribution profiles, making them more attractive for both liver and lungs, whereas non-functionalized NPs were predominantly distributed to the spleen. Formulation NPphe-Rh remained in the brain for at least 2 h after administration.

5.
Pharmaceutics ; 14(5)2022 May 17.
Article in English | MEDLINE | ID: mdl-35631665

ABSTRACT

To date there is no cure for Parkinson's disease (PD), a devastating neurodegenerative disorder with levodopa being the cornerstone of its treatment. In early PD, levodopa provides a smooth clinical response, but after long-term therapy many patients develop motor complications. Tolcapone (TC) is an effective adjunct in the treatment of PD but has a short elimination half-life. In our work, two new controlled delivery systems of TC consisting of biodegradable PLGA 502 (poly (D,L-lactide-co-glycolide acid) microparticles (MPs) and nanoparticles (NPs) were developed and characterized. Formulations MP-TC4 and NP-TC3 were selected for animal testing. Formulation MP-TC4, prepared with 120 mg TC and 400 mg PLGA 502, exhibited a mean encapsulation efficiency (EE) of 85.13%, and zero-order in vitro release of TC for 30 days, with around 95% of the drug released at this time. Formulation NP-TC3, prepared with 10 mg of TC and 50 mg of PLGA 502, exhibited mean EE of 56.69%, particle size of 182 nm, and controlled the release of TC for 8 days. Daily i.p. (intraperitoneal) doses of rotenone (RT, 2 mg/kg) were given to Wistar rats to induce neurodegeneration. Once established, animals received TC in saline (3 mg/kg/day) or encapsulated within formulations MP-TC4 (amount of MPs equivalent to 3 mg/kg/day TC every 14 days) and NP-TC3 (amount of NPs equivalent to 3 mg/kg/day TC every 3 days). Brain analyses of Nissl-staining, GFAP (glial fibrillary acidic protein), and TH (tyrosine hydroxylase) immunohistochemistry as well as behavioral testing (catalepsy, akinesia, swim test) showed that the best formulation was NP-TC3, which was able to revert PD-like symptoms of neurodegeneration in the animal model assayed.

6.
Front Endocrinol (Lausanne) ; 13: 873301, 2022.
Article in English | MEDLINE | ID: mdl-35615716

ABSTRACT

Several neurological diseases share pathological alterations, even though they differ in their etiology. Neuroinflammation, altered brain glucose metabolism, oxidative stress, mitochondrial dysfunction and amyloidosis are biological events found in those neurological disorders. Altered insulin-mediated signaling and brain glucose hypometabolism are characteristic signs observed in the brains of patients with certain neurological diseases, but also others such as type 2 diabetes mellitus and vascular diseases. Thus, significant reductions in insulin receptor autophosphorylation and Akt kinase activity, and increased GSK-3 activity and insulin resistance, have been reported in these neurological diseases as contributing to the decline in cognitive function. Supporting this relationship is the fact that nasal and hippocampal insulin administration has been found to improve cognitive function. Additionally, brain glucose hypometabolism precedes the unmistakable clinical manifestations of some of these diseases by years, which may become a useful early biomarker. Deficiencies in the major pathways of oxidative energy metabolism have been reported in patients with several of these neurological diseases, which supports the hypothesis of their metabolic background. This review remarks on the significance of insulin and brain glucose metabolism alterations as keystone common pathogenic substrates for certain neurological diseases, highlighting new potential targets.


Subject(s)
Brain , Insulin Resistance , Nervous System Diseases , Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Insulin/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Signal Transduction/physiology
7.
Metab Brain Dis ; 36(8): 2597-2602, 2021 12.
Article in English | MEDLINE | ID: mdl-34570340

ABSTRACT

Status epilepticus (SE) is a clinical emergency with high mortality. SE can trigger neuronal death or injury and alteration of neuronal networks resulting in long-term cognitive decline or epilepsy. Among the multiple factors contributing to this damage, imbalance between oxygen and glucose requirements and brain perfusion during SE has been proposed. Herein, we aimed to quantify by neuroimaging the spatiotemporal course of brain perfusion during and after lithium-pilocarpine-induced SE in rats. To this purpose, animals underwent 99mTc-HMPAO SPECT imaging at different time points during and after SE using a small animal SPECT/CT system. 99mTc-HMPAO regional uptake was normalized to the injected dose. In addition, voxel-based statistical parametric mapping was performed. SPECT imaging showed an increase of cortical perfusion before clinical seizure activity onset followed by regional hypo-perfusion starting with the first convulsive seizure and during SE. Twenty-four hours after SE, brain 99mTc-HMPAO uptake was widely decreased. Finally, chronic epileptic animals showed regionally decreased perfusion affecting hippocampus and cortical sub-regions. Despite elevated energy and oxygen requirements, brain hypo-perfusion is present during SE. Our results suggest that insufficient compensation of required blood flow might contribute to neuronal damage and neuroinflammation, and ultimately to chronic epilepsy generated by SE.


Subject(s)
Status Epilepticus , Tomography, Emission-Computed, Single-Photon , Animals , Brain/blood supply , Brain/diagnostic imaging , Cerebrovascular Circulation/physiology , Neuroimaging , Rats , Status Epilepticus/chemically induced , Status Epilepticus/diagnostic imaging , Technetium Tc 99m Exametazime , Tomography, Emission-Computed, Single-Photon/methods
8.
Neuroscience ; 409: 101-110, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31034972

ABSTRACT

Glucose metabolism and serotonergic neurotransmission have been reported to play an important role in epileptogenesis. We therefore aimed to use neuroimaging to evaluate potential alterations in serotonin 5-HT1A receptor and glucose metabolism during epileptogenesis in the rat electrical kindling model. To achieve this goal, we performed positron emission tomography (PET) imaging in a rat epileptogenesis model triggered by electrical stimulation of the hippocampus using 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG), a radiolabeled analog of glucose, and 2'-methoxyphenyl-(N-2'-pyridinyl)-p-18F-fluoro-benzamidoethylpiperazine (18F-MPPF), a radiolabeled 5-HT1A receptor ligand, to evaluate brain metabolism and 5-HT1A receptor functionality. Since the 5-HT1A receptor is also highly expressed in astrocytes, glial fibrillary acidic protein (GFAP) immunofluorescence was performed to detect astrogliosis arising from the kindling procedure once the study was finalized. Lastly, in vitro18F-MPPF autoradiography was performed to evaluate changes in 5HT1A receptor expression. 18F-FDG PET showed reduction of glucose uptake in cortical structures, whereas 18F-MPPF PET revealed an enhancement of tracer binding potential (BPND) in key areas rich in 5-HT1A receptor involved in epilepsy, including septum, hippocampus and entorhinal cortex of kindled animals compared to controls. However, in vitro 5-HT1A receptor autoradiography showed no changes in densitometric signal in any brain region, suggesting that the augmentation in BPND found by PET could be caused by reduction of synaptic serotonin. Importantly, astroglial activation was detected in the hippocampus of kindled rats. Overall, electrical kindling induced hypometabolism, astrogliosis and serotonergic alterations in epilepsy-related regions. Furthermore, the present findings point to 5-HT1A receptor as a valuable epileptogenesis biomarker candidate and a potential therapeutic target.


Subject(s)
Epilepsy/diagnostic imaging , Hippocampus/diagnostic imaging , Kindling, Neurologic/metabolism , Positron-Emission Tomography , Serotonin/metabolism , Animals , Epilepsy/metabolism , Fluorodeoxyglucose F18 , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , Hippocampus/metabolism , Male , Neuroimaging , Rats
9.
Mol Pharm ; 16(5): 1999-2010, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30865462

ABSTRACT

Diseases affecting the central nervous system (CNS) should be regarded as a major health challenge due to the current lack of effective treatments given the hindrance to brain drug delivery imposed by the blood-brain barrier (BBB). Since efficient brain drug delivery should not solely rely on passive targeting, active targeting of nanomedicines into the CNS is being explored. The present study is devoted to the development of lipid nanocapsules (LNCs) decorated with nonpsychotropic cannabinoids as pioneering nonimmunogenic brain-targeting molecules and to the evaluation of their brain-targeting ability both in vitro and in vivo. Noticeably, both the permeability experiments across the hCMEC/D3 cell-based in vitro BBB model and the biodistribution experiments in mice consistently demonstrated that the highest brain-targeting ability was achieved with the smallest-sized cannabinoid-decorated LNCs. Importantly, the enhancement in brain targeting achieved with the conjugation of cannabidiol to LNCs outperformed by 6-fold the enhancement observed for the G-Technology (the main brain active strategy that has already entered clinical trials for the treatment of CNS diseases). As the transport efficiency across the BBB certainly determines the efficacy of the treatments for brain disorders, small cannabinoid-decorated LNCs represent auspicious platforms for the design and development of novel therapies for CNS diseases.


Subject(s)
Blood-Brain Barrier/drug effects , Cannabidiol/pharmacology , Drug Delivery Systems/methods , Lipids/chemistry , Nanocapsules/chemistry , Nanoconjugates/chemistry , Animals , Brain Diseases/drug therapy , Cannabidiol/chemistry , Cannabidiol/metabolism , Capillary Permeability/drug effects , Cell Line , Cell Survival/drug effects , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Humans , Male , Mice , Mice, Inbred ICR , Nanomedicine/methods , Tissue Distribution
10.
Neurochem Int ; 113: 92-106, 2018 02.
Article in English | MEDLINE | ID: mdl-29203398

ABSTRACT

Intracerebral administration of the potassium channel blocker 4-aminopyridine (4-AP) triggers neuronal depolarization and intense acute seizure activity followed by neuronal damage. We have recently shown that, in the lithium-pilocarpine rat model of status epilepticus (SE), a single administration of metyrapone, an inhibitor of the 11ß-hydroxylase enzyme, had protective properties of preventive nature against signs of brain damage and neuroinflammation. Herein, our aim was to investigate to which extent, pretreatment with metyrapone (150 mg/kg, i.p.) was also able to prevent eventual changes in the acute brain metabolism and short-term neuronal damage induced by intrahippocampal injection of 4-AP (7 µg/5 µl). To this end, regional brain metabolism was assessed by 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG) positron emission tomography (PET) during the ictal period. Three days later, markers of neuronal death and hippocampal integrity and apoptosis (Nissl staining, NeuN and active caspase-3 immunohistochemistry), neurodegeneration (Fluoro-Jade C labeling), astrogliosis (glial fibrillary acidic protein (GFAP) immunohistochemistry) and microglia-mediated neuroinflammation (in vitro [18F]GE180 autoradiography) were evaluated. 4-AP administration acutely triggered marked brain hypermetabolism within and around the site of injection as well as short-term signs of brain damage and inflammation. Most important, metyrapone pretreatment was able to reduce ictal hypermetabolism as well as all the markers of brain damage except microglia-mediated neuroinflammation. Overall, our study corroborates the neuroprotective effects of metyrapone against multiple signs of brain damage caused by seizures triggered by 4-AP. Ultimately, our data add up to the consistent protective effect of metyrapone pretreatment reported in other models of neurological disorders of different etiology.


Subject(s)
4-Aminopyridine/toxicity , Glucose/metabolism , Hippocampus/metabolism , Hypoxia, Brain/metabolism , Hypoxia, Brain/prevention & control , Metyrapone/therapeutic use , 4-Aminopyridine/administration & dosage , Animals , Antimetabolites/pharmacology , Antimetabolites/therapeutic use , Glucose/antagonists & inhibitors , Hippocampus/diagnostic imaging , Hippocampus/drug effects , Hypoxia, Brain/chemically induced , Hypoxia, Brain/diagnostic imaging , Injections, Intraventricular , Male , Metyrapone/pharmacology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Positron-Emission Tomography/methods , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/toxicity , Rats , Rats, Sprague-Dawley
11.
Drug Deliv ; 24(1): 1112-1123, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28782388

ABSTRACT

A new drug delivery system is developed for ropinirole (RP) for the treatment of Parkinson's disease (PD) consisting of biodegradable poly (D,L-lactide-co-glycolide) (PLGA) nanoparticles (NPs). The formulation selected was prepared with 8 mg RP and 50 mg PLGA 502. This formulation exhibited mean encapsulation efficiency of 74.8 ± 8.2%, mean particle size lower than 155 nm, the zeta potential of -14.25 ± 0.43 mV and zero-order in vitro release of RP (14.13 ± 0.17 µg/h/10 mg NPs) for 5 d. Daily doses of the neurotoxin rotenone (2 mg/kg) given i.p. to male Wistar rats induced neuronal and behavioral changes similar to those of PD. Once neurodegeneration was established (15 d) animals received RP in saline (1 mg/kg/d for 35 d) or encapsulated within PLGA NPs (amount of NPs equivalent to 1 mg/kg/d RP every 3 d for 35 d). Brain histology and immunochemistry (Nissl-staining, glial fibrillary acidic protein and tyrosine hydroxylase immunohistochemistry) and behavioral testing (catalepsy, akinesia, rotarod and swim test) showed that RP-loaded PLGA NPs were able to revert PD-like symptoms of neurodegeneration in the animal model assayed.


Subject(s)
Nanoparticles , Animals , Drug Carriers , Drug Delivery Systems , Indoles , Lactic Acid , Male , Parkinson Disease , Particle Size , Polyglycolic Acid , Rats , Rats, Wistar
12.
Neuropharmacology ; 123: 261-273, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28495374

ABSTRACT

The status epilepticus (SE) induced by lithium-pilocarpine is a well characterized rodent model of the human temporal lobe epilepsy (TLE) which is accompanied by severe brain damage. Stress and glucocorticoids markedly contribute to exacerbate neuronal damage induced by seizures but the underlying mechanisms are poorly understood. Herein we sought to investigate whether a single administration of metyrapone (150 mg/kg, i.p.), an 11ß-hydroxylase inhibitor, enzyme involved in the peripheral and central synthesis of corticosteroids, had neuroprotective properties in this model. Two experiments were carried out. In exp. 1, metyrapone was administered 3 h before pilocarpine injection whereas in exp. 2, metyrapone administration took place at the onset of the SE. In both experiments, 3 days after the insult, brain metabolism was assessed by in vivo 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG) positron emission tomography (PET). Brains were processed for analyses of markers of hippocampal integrity (Nissl staining), neurodegeneration (Fluoro-Jade C), astrogliosis (glial fibrillary acidic protein (GFAP) immunohistochemistry) and, for a marker of activated microglia by in vitro autoradiography with the TSPO (18 kDa translocator protein) radioligand [18F]GE180. The SE resulted in a consistent hypometabolism in hippocampus, cortex and striatum and neuronal damage, hippocampal neurodegeneration, neuronal death and gliosis. Interestingly, metyrapone had neuroprotective effects when administered before, but not after the insult. In summary, we conclude that metyrapone administration prior but not after the SE protected from brain damage induced by SE in the lithium-pilocarpine model. Therefore, it seems that the effect of metyrapone is preventive in nature and likely related to its antiseizure properties.


Subject(s)
Brain/drug effects , Metyrapone/pharmacology , Neuroprotective Agents/pharmacology , Status Epilepticus/drug therapy , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Autoradiography , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Carbazoles , Carrier Proteins/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Fluorodeoxyglucose F18 , Gliosis/drug therapy , Gliosis/metabolism , Gliosis/pathology , Glucose/metabolism , Immunohistochemistry , Lithium Compounds , Male , Pilocarpine , Positron-Emission Tomography , Radiopharmaceuticals , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Status Epilepticus/diagnostic imaging , Status Epilepticus/metabolism , Status Epilepticus/pathology
13.
Int J Nanomedicine ; 12: 1959-1968, 2017.
Article in English | MEDLINE | ID: mdl-28331318

ABSTRACT

A new nanocarrier is developed for the passage of gatifloxacin through the blood-brain barrier to treat central nervous system tuberculosis. Gatifloxacin nanoparticles were prepared by nanoprecipitation using poly(lactic-co-glycolic acid) (PLGA) 502 and polysorbate 80 or Labrafil as surface modifiers. The evaluation of in vivo blood-brain barrier transport was carried out in male Wistar rats using rhodamine-loaded PLGA nanoparticles prepared with and without the surface modifiers. At 30 and 60 minutes after administration, nanoparticle biodistribution into the brain (hippocampus and cortex), lungs, and liver was studied. The results obtained from the cerebral cortex and hippocampus showed that functionalization of rhodamine nanoparticles significantly increased their passage into the central nervous system. At 60 minutes, rhodamine concentrations decreased in both the lungs and the liver but were still high in the cerebral cortex. To distinguish the effect between the surfactants, gatifloxacin-loaded PLGA nanoparticles were prepared. The best results corresponded to the formulation prepared with polysorbate 80 with regard to encapsulation efficiency (28.2%), particle size (176.5 nm), and ζ-potential (-20.1 mV), thereby resulting in a promising drug delivery system to treat cerebral tuberculosis.


Subject(s)
Fluoroquinolones/therapeutic use , Nanoparticles/chemistry , Tuberculosis, Central Nervous System/drug therapy , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Chemistry, Pharmaceutical , Fluoroquinolones/pharmacology , Gatifloxacin , Male , Microscopy, Confocal , Neurons/drug effects , Neurons/pathology , Rats, Wistar , Rhodamines/metabolism , Solutions , Tissue Distribution/drug effects
14.
Pharmacol Biochem Behav ; 153: 32-44, 2017 02.
Article in English | MEDLINE | ID: mdl-27988176

ABSTRACT

Fluoxetine (FLX) has paradoxical anxiogenic-like effects during the acute phase of treatment. In adolescent (35d-old) male rats, the stress-like effects induced by short-term (3d-4d) FLX treatment appear to involve up-regulation of paraventricular nucleus (PVN) arginine vasopressin (AVP) mRNA. However, studies on FLX-induced anxiety-like effects in adult rodents are inconclusive. Herein, we sought to study the response of adult male rats (60-65d-old) to a similar FLX treatment, also investigating how the stressful component, inherent to our experimental conditions, contributed to the responses. We show that FLX acutely increased plasma corticosterone concentrations while it attenuated the stress-induced-hyperthermia (SIH) as well as it reduced (≈40%) basal POMC mRNA expression in the arcuate nucleus (ARC). However, FLX did not alter the basal expression of PVN-corticotrophin-releasing hormone (CRH), anterior pituitary-pro-opiomelanocortin (POMC) and raphe nucleusserotonin transporter (SERT). Nonetheless, some regressions point towards the plausibility that FLX activated the hypothalamic-pituitary-adrenal (HPA). The behavioral study revealed that FLX acutely increased emotional reactivity in the holeboard, effect followed by a body weight loss of ≈2.5% after 24h. Interestingly, i.p. injection with vehicle did not have behavioral effects, furthermore, after experiencing the stressful component of the holeboard, the rats kept eating and gaining weight as normal. By contrast, the stress-naïve rats reduced food intake and gained less weight although maintaining a positive energy state. Therefore, on one hand, repetition of a mild stressor would unchain compensatory mechanisms to restore energy homeostasis after stress increasing the resiliency to novel stressors. On the other hand, FLX might render stressed adult rats vulnerable to novel stressors through the emergence of counter-regulatory changes, involving HPA axis activation and diminished sympathetic output may be due to reduced melanocortin signaling. Therefore, complex interactions between hypothalamic CRH and POMC might be determining the adaptive nature of the response of adult male rats to FLX and/or stress.


Subject(s)
Anxiety/chemically induced , Fluoxetine/pharmacology , Stress, Psychological/etiology , Animals , Corticosterone/blood , Corticotropin-Releasing Hormone/physiology , Energy Metabolism/drug effects , Hypothalamo-Hypophyseal System/physiology , Hypothalamus/physiology , Male , Pituitary-Adrenal System/physiology , Pro-Opiomelanocortin/physiology , Rats , Rats, Sprague-Dawley , Serotonin Plasma Membrane Transport Proteins/genetics
15.
CNS Neurol Disord Drug Targets ; 16(6): 694-704, 2017.
Article in English | MEDLINE | ID: mdl-27989232

ABSTRACT

BACKGROUND: Epilepsy is a central disorder associated with neuronal damage and brain hypometabolism. It has been reported that antidepressant drugs show anticonvulsant and neuroprotective effects in different animal models of seizures and epilepsy. AIMS: The purpose of this study was to investigate the eventual short-term brain impairment induced by a single low convulsant dose of the potassium channel blocker 4-aminopyridine (4-AP) and the eventual neuroprotective effects exerted by fluoxetine, a prototypical selective serotonin (5-hydroxytryptamine; 5-HT) reuptake inhibitor (SSRI). METHOD: In vivo 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET) and several histological assessments were carried out in adult male rats after i.p. administration of 3 mg/kg 4-AP for evaluating eventual brain metabolism impairment and signs of hippocampal damage. We also evaluated the effects of a short-term fluoxetine treatment (10 mg/kg, i.p. for 7 days) in this seizure model. RESULTS: [18F]FDG PET analysis revealed no changes in the regional brain metabolism on day 3 after 4-AP injection. The histological assessments revealed signs of damage in the hippocampus, a brain area usually affected by seizures. Thus, reactive gliosis and a significant increase in the expression of caspase-9 were found in the aforementioned brain area. By contrast, we observed no signs of neurodegeneration or neuronal death. Regarding the effects of fluoxetine, this SSRI showed beneficial neurologic effects, since it significantly increased the seizure latency time and reduced the abovementioned 4-AP-induced hippocampal damage markers. CONCLUSION: Overall, our results point to SSRIs and eventually endogenous 5-HT as neuroprotective agents against convulsant-induced hippocampal damage.


Subject(s)
4-Aminopyridine/toxicity , Brain Injuries , Convulsants/toxicity , Fluoxetine/therapeutic use , Hippocampus/pathology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Animals , Brain Injuries/chemically induced , Brain Injuries/drug therapy , Brain Injuries/pathology , Caspase 9/metabolism , Cell Death/drug effects , Disease Models, Animal , Fluoresceins/metabolism , Fluorodeoxyglucose F18/pharmacokinetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/chemically induced , Gliosis/pathology , Hippocampus/diagnostic imaging , Hippocampus/drug effects , Male , Phosphopyruvate Hydratase/metabolism , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects
16.
Curr Pharm Des ; 23(23): 3423-3431, 2017.
Article in English | MEDLINE | ID: mdl-27779080

ABSTRACT

A new controlled delivery system has been developed for ropinirole (RP) for the treatment of Parkinson´s Disease (PD) consisting in PLGA microparticles (MPs) which exhibited in vitro constant release of RP (78.23 µg/day/10 mg MPs) for 19 days. The neuroprotective effects of RP released from MPs were evaluated in SKN-AS cells after exposure to rotenone (20 µM). Cell apoptosis was significantly reduced by RP (100-120 µM). Daily doses of rotenone (2 mg/kg) given i.p. to rats induced neuronal and behavioral changes similar to those of PD. After 15 days, animals received RP in saline (1 mg/kg/day for 45 days) or as MPs at two dose levels (amount of MPs equivalent to 7.5 mg/kg or 15 mg/kg RP given on days 15 and 30). Brain immunochemistry (Nisslstaining, GFAP and TH immunohistochemistry) and behavioral testing (catalepsy, akinesia, rotarod and swim test) showed that animals receiving RP either in solution or encapsulated within the MPs reverted the PD symptoms with the best results obtained in animals receiving RP microspheres at the highest dose assayed, thereby confirming the potential therapeutic interest of the new RP delivery system.


Subject(s)
Antiparkinson Agents/administration & dosage , Indoles/administration & dosage , Lactic Acid/administration & dosage , Microspheres , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Polyglycolic Acid/administration & dosage , Rotenone/toxicity , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Humans , Insecticides/toxicity , Male , Parkinsonian Disorders/pathology , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Wistar , Treatment Outcome
17.
Diabetes ; 66(1): 64-74, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27999108

ABSTRACT

Brain activity requires a flux of glucose to active regions to sustain increased metabolic demands. Insulin, the main regulator of glucose handling in the body, has been traditionally considered not to intervene in this process. However, we now report that insulin modulates brain glucose metabolism by acting on astrocytes in concert with IGF-I. The cooperation of insulin and IGF-I is needed to recover neuronal activity after hypoglycemia. Analysis of underlying mechanisms show that the combined action of IGF-I and insulin synergistically stimulates a mitogen-activated protein kinase/protein kinase D pathway resulting in translocation of GLUT1 to the cell membrane through multiple protein-protein interactions involving the scaffolding protein GAIP-interacting protein C terminus and the GTPase RAC1. Our observations identify insulin-like peptides as physiological modulators of brain glucose handling, providing further support to consider the brain as a target organ in diabetes.


Subject(s)
Astrocytes/metabolism , Glucose/metabolism , Animals , Biological Transport/physiology , Glucose Transporter Type 1/metabolism , Glycogen/metabolism , Immunoassay , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Lactic Acid/metabolism , Male , Mice , Neurons/metabolism , Plasmids , Polymerase Chain Reaction , Positron-Emission Tomography
18.
Neuroscience ; 339: 191-209, 2016 Dec 17.
Article in English | MEDLINE | ID: mdl-27717809

ABSTRACT

Spinocerebellar ataxia type-3 (SCA-3) is a rare disease but it is the most frequent type within the autosomal dominant inherited ataxias. The disease lacks an effective treatment to alleviate major symptoms and to modify disease progression. Our recent findings that endocannabinoid receptors and enzymes are significantly altered in the post-mortem cerebellum of patients affected by autosomal-dominant hereditary ataxias suggest that targeting the endocannabinoid signaling system may be a promising therapeutic option. Our goal was to investigate the status of the endocannabinoid signaling system in a transgenic mouse model of SCA-3, in the two CNS structures most affected in this disease - cerebellum and brainstem. These animals exhibited progressive motor incoordination, imbalance, abnormal gait, muscle weakness, and dystonia, in parallel to reduced in vivo brain glucose metabolism, deterioration of specific neuron subsets located in the dentate nucleus and pontine nuclei, small changes in microglial morphology, and reduction in glial glutamate transporters. Concerning the endocannabinoid signaling, our data indicated no changes in CB2 receptors. By contrast, CB1 receptors increased in the Purkinje cell layer, in particular in terminals of basket cells, but they were reduced in the dentate nucleus. We also measured the levels of endocannabinoid lipids and found reductions in anandamide and oleoylethanolamide in the brainstem. These changes correlated with an increase in the FAAH enzyme in the brainstem, which also occurred in some cerebellar areas, whereas other endocannabinoid-related enzymes were not altered. Collectively, our results in SCA-3 mutant mice confirm a possible dysregulation in the endocannabinoid system in the most important brain structures affected in this type of ataxia, suggesting that a pharmacological manipulation addressed to correct these changes could be a promising option in SCA-3.


Subject(s)
Brain Stem/metabolism , Cerebellum/metabolism , Endocannabinoids/metabolism , Machado-Joseph Disease/metabolism , Amidohydrolases/metabolism , Animals , Brain Stem/diagnostic imaging , Brain Stem/pathology , Cerebellum/diagnostic imaging , Cerebellum/pathology , Disease Models, Animal , Glucose/metabolism , Machado-Joseph Disease/pathology , Mice, Transgenic , Motor Activity/physiology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Phenotype , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism
19.
Mol Imaging Biol ; 18(5): 733-40, 2016 10.
Article in English | MEDLINE | ID: mdl-27043223

ABSTRACT

PURPOSE: Epileptogenesis, i.e., development of epilepsy, involves a number of processes that alter the brain function in the way that triggers spontaneous seizures. Kindling is one of the most used animal models of temporal lobe epilepsy (TLE) and epileptogenesis, although chemical kindling suffers from high inter-assay success unpredictability. This study was aimed to analyze the eventual regional brain metabolic changes during epileptogenesis in the pentylenetetrazole (PTZ) kindling model in order to obtain a predictive kindling outcome parameter. PROCEDURES: In vivo longitudinal positron emission tomography (PET) scans with 2-deoxy-2-[(18)F]fluoro-D-glucose ([(18)F]FDG) along the PTZ kindling protocol (35 mg/kg intraperitoneally (i.p.), 18 sessions) in adult male rats were performed in order to evaluate the regional brain metabolism. RESULTS: The half of the PTZ-injected rats reached the kindled state. In addition, a significant decrease of [(18)F]FDG uptake at the end of the protocol in most of the brain structures of kindled animals was found, reflecting the characteristic epilepsy-associated hypometabolism. However, PTZ-injected animals but not reaching the kindled state did not show this widespread brain hypometabolism. Retrospective analysis of the data revealed that hippocampal [(18)F]FDG uptake normalized to pons turned out to be a predictive index of the kindling outcome. Thus, a 19.06 % reduction (p = 0.008) of the above parameter was found in positively kindled rats compared to non-kindled ones just after the fifth PTZ session. CONCLUSION: Non-invasive PET neuroimaging was a useful tool for discerning epileptogenesis progression in this animal model. Particularly, the [(18)F]FDG uptake of the hippocampus proved to be an early predictive parameter to differentiate resistant and non-resistant animals to the PTZ kindling.


Subject(s)
Fluorodeoxyglucose F18/chemistry , Kindling, Neurologic/pathology , Neuroimaging/methods , Positron-Emission Tomography/methods , Animals , Disease Progression , Magnetic Resonance Imaging , Male , Pentylenetetrazole , Rats, Sprague-Dawley , Seizures/diagnostic imaging , Seizures/pathology
20.
Neuropharmacology ; 110(Pt A): 519-529, 2016 11.
Article in English | MEDLINE | ID: mdl-26976670

ABSTRACT

Cannabinoid CB2 receptors (CB2Rs) are emerging as important therapeutic targets in brain disorders that typically involve neurometabolic alterations. We here addressed the possible role of CB2Rs in the regulation of glucose uptake in the mouse brain. To that aim, we have undertaken 1) measurement of (3)H-deoxyglucose uptake in cultured cortical astrocytes and neurons and in acute hippocampal slices; 2) real-time visualization of fluorescently labeled deoxyglucose uptake in superfused hippocampal slices; and 3) in vivo PET imaging of cerebral (18)F-fluorodeoxyglucose uptake. We now show that both selective (JWH133 and GP1a) as well as non-selective (WIN55212-2) CB2R agonists, but not the CB1R-selective agonist, ACEA, stimulate glucose uptake, in a manner that is sensitive to the CB2R-selective antagonist, AM630. Glucose uptake is stimulated in astrocytes and neurons in culture, in acute hippocampal slices, in different brain areas of young adult male C57Bl/6j and CD-1 mice, as well as in middle-aged C57Bl/6j mice. Among the endocannabinoid metabolizing enzymes, the selective inhibition of COX-2, rather than that of FAAH, MAGL or α,ßDH6/12, also stimulates the uptake of glucose in hippocampal slices of middle-aged mice, an effect that was again prevented by AM630. However, we found the levels of the endocannabinoid, anandamide reduced in the hippocampus of TgAPP-2576 mice (a model of ß-amyloidosis), and likely as a consequence, COX-2 inhibition failed to stimulate glucose uptake in these mice. Together, these results reveal a novel general glucoregulatory role for CB2Rs in the brain, raising therapeutic interest in CB2R agonists as nootropic agents.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Glucose/metabolism , Receptor, Cannabinoid, CB2/metabolism , Aging/drug effects , Aging/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor , Amyloidosis/diagnostic imaging , Amyloidosis/drug therapy , Amyloidosis/metabolism , Animals , Arachidonic Acids/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Brain/diagnostic imaging , Brain/drug effects , Cannabinoid Receptor Modulators/pharmacology , Cells, Cultured , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Endocannabinoids/metabolism , Hydroxyethylrutoside , Male , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Nootropic Agents/pharmacology , Polyunsaturated Alkamides/metabolism , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...