Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 12.075
1.
N Engl J Med ; 389(4): 309-321, 2023 Jul 27.
Article En | MEDLINE | ID: mdl-37494485

BACKGROUND: Narcolepsy type 1 is caused by severe loss or lack of brain orexin neuropeptides. METHODS: We conducted a phase 2, randomized, placebo-controlled trial of TAK-994, an oral orexin receptor 2-selective agonist, in patients with narcolepsy type 1. Patients with confirmed narcolepsy type 1 according to clinical criteria were randomly assigned to receive twice-daily oral TAK-994 (30 mg, 90 mg, or 180 mg) or placebo. The primary end point was the mean change from baseline to week 8 in average sleep latency (the time it takes to fall asleep) on the Maintenance of Wakefulness Test (range, 0 to 40 minutes; normal ability to stay awake, ≥20 minutes). Secondary end points included the change in the Epworth Sleepiness Scale (ESS) score (range, 0 to 24, with higher scores indicating greater daytime sleepiness; normal, <10) and the weekly cataplexy rate. RESULTS: Of the 73 patients, 17 received TAK-994 at a dose of 30 mg twice daily, 20 received 90 mg twice daily, 19 received 180 mg twice daily, and 17 received placebo. The phase 2 trial and an extension trial were terminated early owing to hepatic adverse events. Primary end-point data were available for 41 patients (56%); the main reason for missing data was early trial termination. Least-squares mean changes to week 8 in average sleep latency on the MWT were 23.9 minutes in the 30-mg group, 27.4 minutes in the 90-mg group, 32.6 minutes in the 180-mg group, and -2.5 minutes in the placebo group (difference vs. placebo, 26.4 minutes in the 30-mg group, 29.9 minutes in the 90-mg group, and 35.0 minutes the 180-mg group; P<0.001 for all comparisons). Least-squares mean changes to week 8 in the ESS score were -12.2 in the 30-mg group, -13.5 in the 90-mg group, -15.1 in the 180-mg group, and -2.1 in the placebo group (difference vs. placebo, -10.1 in the 30-mg group, -11.4 in the 90-mg group, and -13.0 in the 180-mg group). Weekly incidences of cataplexy at week 8 were 0.27 in the 30-mg group, 1.14 in the 90-mg group, 0.88 in the 180-mg group, and 5.83 in the placebo group (rate ratio vs. placebo, 0.05 in the 30-mg group, 0.20 in the 90-mg group, and 0.15 in the 180-mg group). A total of 44 of 56 patients (79%) receiving TAK-994 had adverse events, most commonly urinary urgency or frequency. Clinically important elevations in liver-enzyme levels occurred in 5 patients, and drug-induced liver injury meeting Hy's law criteria occurred in 3 patients. CONCLUSIONS: In a phase 2 trial involving patients with narcolepsy type 1, an orexin receptor 2 agonist resulted in greater improvements on measures of sleepiness and cataplexy than placebo over a period of 8 weeks but was associated with hepatotoxic effects. (Funded by Takeda Development Center Americas; TAK-994-1501 and TAK-994-1504 ClinicalTrials.gov numbers, NCT04096560 and NCT04820842.).


Narcolepsy , Orexin Receptors , Orexins , Humans , Cataplexy/complications , Cataplexy/drug therapy , Cataplexy/epidemiology , Double-Blind Method , Narcolepsy/drug therapy , Narcolepsy/complications , Narcolepsy/epidemiology , Orexin Receptors/agonists , Orexin Receptors/therapeutic use , Sleepiness/drug effects , Treatment Outcome , Orexins/analysis , Orexins/deficiency , Orexins/pharmacology , Brain Chemistry/drug effects , Administration, Oral , Chemical and Drug Induced Liver Injury/etiology
2.
Sci Rep ; 12(1): 996, 2022 01 19.
Article En | MEDLINE | ID: mdl-35046433

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of extracellular amyloid-beta peptides (Aß) resulting in senile plaques and intracellular hyperphosphorylated tau protein resulting in neurofibrillary tangles (NFTs). Mucuna beans (Mucuna pruriences (L.) DC. var. utilis) are unique plants containing 3-9% L-3,4-dihydroxyphenylalanine (L-DOPA). Here we investigated the effect of the administration of Mucuna beans on AD prevention by feeding triple-transgenic mice (3 × Tg-AD mice) with a diet containing Mucuna beans for 13 months. The levels of Aß oligomers and detergent-insoluble phosphorylated tau decreased in the brain of mice fed with Mucuna beans (Mucuna group) compared to those of the Control group. Aß accumulation and phosphorylated tau accumulation in the brain in the Mucuna group were also reduced. In addition, administration of Mucuna beans improved cognitive function. These results suggest that administration of Mucuna beans may have a preventive effect on AD development in 3 × Tg-AD mice.


Alzheimer Disease/drug therapy , Brain Chemistry/drug effects , Mucuna/chemistry , Alzheimer Disease/genetics , Amyloid beta-Peptides/analysis , Animals , Cognition/drug effects , Diet/veterinary , Disease Models, Animal , Female , Levodopa/analysis , Mice, Transgenic , tau Proteins/analysis
3.
J Neurochem ; 160(2): 271-282, 2022 01.
Article En | MEDLINE | ID: mdl-34699608

Alcohol-related brain injury is characterized by cognitive deficits and brain atrophy with the prefrontal cortex particularly susceptible. White matter in the human brain is lipid rich and a major target of damage from chronic alcohol abuse; yet, there is sparse information on how these lipids are affected. Here, we used untargeted lipidomics as a discovery tool to describe these changes in the prefrontal, middle temporal, and visual cortices of human subjects with alcohol use disorder and controls. Significant changes to the lipidome, predominantly in the prefrontal and visual cortices, and differences between the white and grey matter of each brain region were identified. These effects include broad decreases to phospholipids and ceramide, decreased polyunsaturated fatty acids, decreased sphingadiene backbones, and selective decreases in cholesteryl ester fatty acid chains. Our findings show that chronic alcohol abuse results in selective changes to the neurolipidome, which likely reflects both the directs effects on the brain and concurrent effects on the liver.


Alcoholism/complications , Brain Chemistry/drug effects , Brain/drug effects , Brain/metabolism , Lipidomics , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged
4.
Neurotoxicology ; 88: 106-115, 2022 01.
Article En | MEDLINE | ID: mdl-34793780

Lead (Pb) is a well-known neurotoxicant and environmental hazard. Recent experimental evidence has linked Pb exposure with neurological deterioration leading to neurodegenerative diseases, such as Alzheimer's disease. To understand brain regional distribution of Pb and its interaction with other metal ions, we used synchrotron micro-x-ray fluorescence technique (µ-XRF) to map the metal distribution pattern and to quantify metal concentrations in mouse brains. Lead-exposed mice received oral gavage of Pb acetate once daily for 4 weeks; the control mice received sodium acetate. Brain tissues were cut into slices and subjected for analysis. Synchrotron µ-XRF scans were run on the PETRA III P06 beamline (DESY). Coarse scans of the entire brain were performed to locate the cortex and hippocampus, after which scans with higher resolution were run in these areas. The results showed that: a) the total Pb intensity in Pb-exposed brain slices was significantly higher than in control brain; b) Pb typically deposited in localized particles of <10 um2 in both the Pb-exposed and control brain slices, with more of these particles in Pb-exposed samples; c) selenium (Se) was significantly correlated with Pb in these particles in the cortex and hippocampus/corpus callosum regions in the Pb-exposed samples, and the molar ratio of the Se and Pb in these particles is close to 1:1. These results indicated that Se may play a crucial role in Pb-induced neurotoxicity. Our findings call for further studies to investigate the relationship between Pb exposure and possible Se detoxification responses, and the implication in the etiology of Alzheimer's disease.


Brain Chemistry/drug effects , Lead Poisoning, Nervous System/metabolism , Lead/analysis , Selenium/analysis , Animals , Lead/administration & dosage , Male , Mice , Spectrometry, X-Ray Emission , Synchrotrons
5.
Neurotoxicology ; 88: 65-78, 2022 01.
Article En | MEDLINE | ID: mdl-34742947

Synthetic cathinones are used as stimulants of abuse. Many abused drugs, including stimulants, activate nuclear factor-κB (NF-κB) transcription leading to increases in NF-κB-regulated pro-inflammatory cytokines, and the level of inflammation appears to correlate with length of abuse. The purpose of this study was to measure the profile of IL-1α, IL-1ß, IL-6, CCL2 and TNF-α in brain and plasma to examine if drug exposure alters inflammatory markers. Male and female Sprague-Dawley rats were trained to self-administer α-pyrrolidinopentiophenone (α-PVP) (0.1 mg/kg/infusion), 4-methylmethcathinone (4MMC) (0.5 mg/kg/infusion), or saline through autoshaping, and then self-administered for 21 days during 1 h (short access; ShA) or 6 h (long access; LgA) sessions. Separate rats were assigned to a naïve control group. Cytokine levels were examined in amygdala, hippocampus, hypothalamus, prefrontal cortex, striatum, thalamus, and plasma. Rats acquired synthetic cathinone self-administration, and there were no sex differences in drug intake. Synthetic cathinone self-administration produced sex differences in IL-1α, IL-1ß, IL-6, CCL2 and TNF-α levels. There were widespread increases in inflammatory cytokines in the brains of male rats compared to females, particularly for 4MMC, whereas females were more likely to show increased inflammatory cytokines in plasma compared to saline groups than males. Furthermore, these sex differences in cytokine levels were more common after LgA access to synthetic cathinones than ShA. These results suggest that synthetic cathinone use likely produces sex-selective patterns of neuroinflammation during the transition from use to abuse. Consequently, treatment need may differ depending on the progression of synthetic cathinone abuse and based on sex.


Alkaloids/pharmacology , Central Nervous System Stimulants/pharmacology , Cytokines/analysis , Alkaloids/administration & dosage , Animals , Brain Chemistry/drug effects , Central Nervous System Stimulants/administration & dosage , Chemokine CCL2/analysis , Chemokine CCL2/blood , Cytokines/blood , Female , Interleukin-1alpha/analysis , Interleukin-1alpha/blood , Interleukin-1beta/analysis , Interleukin-1beta/blood , Interleukin-6/analysis , Interleukin-6/blood , Male , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/metabolism , Rats , Rats, Sprague-Dawley , Self Administration , Sex Factors , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/blood
6.
Article En | MEDLINE | ID: mdl-34743051

Obesity drives an imbalanced signature of specialized pro-resolving mediators (SPM). Herein, we investigated if high fat diet-induced obesity dysregulates the concentration of SPM intermediates in the brains of C57BL/6 J mice. Furthermore, given the benefits of EPA for cardiometabolic diseases, major depression, and cognition, we probed the effect of an EPA supplemented high fat diet on brain SPM intermediates. Mass spectrometry revealed no effect of the high fat diet on PUFA-derived brain metabolites. EPA also did not have an effect on most brain PUFA-derived metabolites except an increase of 12-hydroxyeicosapentaenoic acid (12-HEPE). In contrast, EPA dramatically increased serum HEPEs and lowered several PUFA-derived metabolites. Finally, untargeted mass spectrometry showed no effects of the high fat diet, with or without EPA, on the brain metabolome. Collectively, these results show the murine brain resists a deficiency in SPM pathway markers in response to a high fat diet and that EPA supplementation increases 12-HEPE levels.


Brain Chemistry/drug effects , Diet, High-Fat/adverse effects , Eicosapentaenoic Acid/analogs & derivatives , Lipoxins/metabolism , Obesity/metabolism , Animals , Disease Models, Animal , Eicosapentaenoic Acid/administration & dosage , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/metabolism , Eicosapentaenoic Acid/pharmacology , Lipoxins/analysis , Male , Mass Spectrometry , Metabolic Networks and Pathways , Metabolomics/methods , Mice , Mice, Obese , Obesity/chemically induced
7.
Food Chem Toxicol ; 158: 112665, 2021 Dec.
Article En | MEDLINE | ID: mdl-34780879

Excessive fluoride is capable of inducing cognitive deficits, but the mechanisms remain elusive. This study aimed to investigate the effects and underlying mechanisms of fluoride on mitochondrial dysfunction and neurobiological alterations, as well as cognitive impairment. C57BL/6 mice were orally administered 25, 50, and 100 mg/L NaF for 90 days. Cultured human neuroblastoma SH-SY5Y cells were exposed to NaF (110 mg/L) for 24 h in the presence or absence of Sirt3 overexpression. The results demonstrated that chronic exposure to high fluoride induced cognitive deficits and neural/synaptic injury in mice. Fluoride reduced mitochondrial antioxidant enzyme activities and elevated SOD2 acetylation by downregulating Sirt3 expression in the brains of mice and NaF-treated SH-SY5Y cells. Moreover, fluoride lowered mtDNA transcription and induced mitochondrial dysfunction along with increased FoxO3A acetylation in the brains of mice and NaF-treated SH-SY5Y cells. Subsequent experiments revealed that overexpression of Sirt3 significantly attenuated the adverse effects of fluoride on radical scavenging capabilities and mtDNA transcription, as well as mitochondrial function in SH-SY5Y cells. These results suggest that chronic long-term fluoride exposure evokes neural/synaptic injury and cognitive impairment through mitochondrial dysfunction and its associated oxidative stress, which is, at least partly, mediated by Sirt3 inhibition in the mouse brain.


Cognitive Dysfunction/chemically induced , Mitochondria , Sirtuin 3 , Sodium Fluoride/toxicity , Animals , Brain Chemistry/drug effects , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Morris Water Maze Test/drug effects , Neurotoxins/toxicity , Sirtuin 3/genetics , Sirtuin 3/metabolism
8.
J Chem Neuroanat ; 118: 102015, 2021 12.
Article En | MEDLINE | ID: mdl-34454018

Oxidative stress, an adverse consequence of brain ischemia-reperfusion injury (IRI), activates matrix metalloproteinase enzymes which cause to destruction of extracellular matrix and tight junction proteins. Oxidative stress during stroke increases serum endothelin-1 and endothelin B receptor (ETBR) expression. Apelin-13, an endogenous peptide, is expressed in numerous tissues that regulate diverse physiological and pathological processes. This study aimed to investigate the effect of intravenous (IV) injection of apelin-13 on cerebral vasogenic edema due to brain IRI. Animals were divided into sham, ischemia, and treat groups. IRI model was induced by middle cerebral artery occlusion (MCAO) for 60 min followed by 23 h reperfusion. Apelin-13 was injected into the tail vein 5 min before reperfusion. Neurological defects were evaluated with longa test. Brain water content and BBB permeability were assessed according to cerebral dry-wet weight and brain Evans blue extraction. Malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT) were measured using the colorimetric method. Expression of occludin and claudin-5, matrix metalloproteinase- 2 and 9 (MMP-2 & 9) and, ETBR were evaluated using Western blot. Brain IRI was associated with BBB breakdowns and vasogenic edema. Apelin-13 significantly reduced BBB permeability and vasogenic edema. Apelin-13 significantly attenuated IRI-related oxidative stress. Apelin-13 decreased expression of mmp-2, 9 and ETBR, prevented from decrement of occludin and claudin-5 expersion, which protected BBB integrity and reduced vasogenic edema. In conclusion, our results have suggested that an IV injection of apelin-13 could somehow reduce vasogenic edema via targeting oxidative stress and ETBR expression.


Claudin-5/antagonists & inhibitors , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/therapeutic use , Ischemic Stroke/drug therapy , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinase Inhibitors/therapeutic use , Occludin/antagonists & inhibitors , Oxidative Stress/drug effects , Receptor, Endothelin B/drug effects , Animals , Antioxidants/metabolism , Brain Chemistry/drug effects , Brain Edema/drug therapy , Infarction, Middle Cerebral Artery/drug therapy , Infusions, Intravenous , Male , Rats , Rats, Wistar
9.
Brain Res Bull ; 175: 234-243, 2021 10.
Article En | MEDLINE | ID: mdl-34333049

INTRODUCTION: The contribution of neuroinflammation in cognitive impairment is increasingly recognized. Non-steroidal anti-inflammatory drugs had been proven that it could improve cognitive impairment in large dose but with more side effect, which limited the application. The main objective of this study was to investigate whether the combined use of nicotine and celecoxib could obtain synergistic neuroprotective effect in ischemic rats. METHODS: Twenty adult Sprague-Dawley (SD) rats underwent ischemic model surgery by injecting endothelin-1 into the left thalamus, which were classified into four groups with different interventions: nicotine (1.5 mg/kg/d), celecoxib (15 mg/kg/d), nicotine (1.5 mg/kg/d) +celecoxib (15 mg/kg/d), or saline after surgery. The other five SD rats also underwent same surgery by injecting saline instead of endothelin-1, as the control group. Morris water maze (MWM) test was adopted to assess the cognition. Micro PET/CT with 2-[18F]-A-85380 were performed for α4ß2-nAChRs detection in vivo. Western blot, real-time PCR and immunohistochemical staining were adopted to detect the expression of α4ß2-nAChRs and inflammatory factors which included TNF-α, IL-1ß, IL-6 in brain tissue. Microglial activation in the brain was monitored by immunofluorescence with IBA1 staining. RESULTS: The MWM test showed rats given with nicotine or celecoxib alone showed much better memory than rats with saline, no difference was observed between nicotine and celecoxib. The rat memory was recovered most significant when the nicotine and celecoxib were combined (p < 0.05). Micro-PET/CT showed much more tracer uptake in the left thalamus and whole brain in rats given with nicotine, or nicotine + celecoxib (nico + cele group) than saline treated rats, whereas the rats given celecoxib did not. Compared with saline treated rats, we found the proteins of α4nAChR and ß2nAChR in rats given nicotine or nico + cele increased significantly, and mRNA/proteins of TNF-α, IL-1ß and IL-6 decreased at the same time. The α 4nAChR and ß 2nAChR proteins in rats given celecoxib is the same as saline treated rats, whereas the inflammatory factors decreased obviously compared with saline treated rats. Microglial activation was confirmed in saline treated rats, which was inhibited in rats give nicotine, celecoxib or both. CONCLUSIONS: The study revealed the combined use of nicotine and celecoxib may improve the cognitive function in ischemic rats, with a better effect than either alone. Both nicotine and celecoxib can inhibit inflammation, but through different mechanisms: nicotine can activate α4ß2-nAChRs while celecoxib is cyclooxygenase-2 inhibitor. Our findings suggest the combined application of two drugs with different anti-inflammation mechanism could attenuate cognitive impairment more effectively in ischemic rats, which may hold therapeutic potential in the clinical practice.


Brain Ischemia/drug therapy , Celecoxib/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Neuroinflammatory Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Nicotine/therapeutic use , Nicotinic Agonists/therapeutic use , Animals , Brain Chemistry/drug effects , Calcium-Binding Proteins/biosynthesis , Cognition/drug effects , Cytokines/biosynthesis , Drug Synergism , Drug Therapy, Combination , Endothelin-1/pharmacology , Male , Maze Learning/drug effects , Microfilament Proteins/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/metabolism , X-Ray Microtomography
10.
Brain Res Bull ; 175: 186-195, 2021 10.
Article En | MEDLINE | ID: mdl-34333052

Puberty includes a highly stress-sensitive period with significant sex differences in the neurophysiological and behavioural outcomes of a peripheral immune challenge. Sex differences in the pubertal neuroimmune network's responses to systemic LPS may explain some of these enduring sex-specific outcomes of a pubertal immune challenge. However, the functional implications of these sex-specific neuroimmune responses on the local microenvironment are unclear. Western blots were used to examine treatment- and sex-related changes in expression of regulatory proteins in inflammation (NFκB), cell death (AIF), oxidative stress (SOD-1), and synaptic plasticity (PSD-95) following symptomatic recovery (i.e., one week post-treatment) from pubertal immune challenge. Across the four examined brain regions (i.e., hippocampus, PFC, hypothalamus, and cerebellum), only PSD-95 levels were altered one week post-treatment by the pubertal LPS treatment. Unlike their female counterparts, seven-week-old males showed increased PSD-95 expression in the hippocampus (p < .05). AIF, SOD-1, and NFκB levels in both sexes were unaffected by treatment (all p > .05), which suggests appropriate resolution of NFκB-mediated immune responses to pubertal LPS without stimulating AIF-mediated apoptosis and oxidative stress. We also report a significant male-biased sex difference in PSD-95 levels in the PFC and in cerebellar expression of SOD-1 during puberty (all p < .05). These findings highlight the sex-specific vulnerability of the pubertal hippocampus to systemic LPS and suggest that a pubertal immune challenge may expedite neurodevelopment in the hippocampus in a sex-specific manner.


Disks Large Homolog 4 Protein/biosynthesis , Lipopolysaccharides/pharmacology , Sexual Maturation , Animals , Apoptosis/drug effects , Apoptosis/genetics , Body Weight/drug effects , Brain Chemistry/drug effects , Brain Chemistry/genetics , Disks Large Homolog 4 Protein/genetics , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , NF-kappa B/metabolism , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Oxidative Stress/drug effects , Oxidative Stress/genetics , Sex Characteristics , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics
11.
Neuropharmacology ; 197: 108713, 2021 10 01.
Article En | MEDLINE | ID: mdl-34271019

Alcohol is the most commonly used psychoactive drug, often taken in conjunction with opioid drugs. Since both alcohol and opioids can induce CNS depression, it is often assumed that alcohol potentiates the known hypoxic effects of opioid drugs. To address this supposition, we used oxygen sensors to examine the effects of alcohol on brain oxygenation and hypoxic responses induced by intravenous heroin in awake, freely moving rats. To eliminate robust sensory effects of alcohol following its oral or intraperitoneal delivery, alcohol was administered directly into the stomach via chronically implanted intragastric catheters at human relevant doses. Alcohol delivered at a 0.5 g/kg dose did not affect brain oxygen levels, except for a weak transient increase during drug delivery. This phasic oxygen increase was stronger at a 2.0 g/kg alcohol dose and followed by a weaker tonic increase. Since alcohol absorption from intragastric delivery is much slower and more prolonged than with intraperitoneal or intravenous injections, the rapid rise of brain oxygen levels suggests that alcohol has a direct action on sensory afferents in the stomach well before the drug physically reaches brain tissue via circulation. Despite slow tonic increases in brain oxygen, alcohol at the 2.0 g/kg dose strongly potentiates heroin-induced oxygen responses, increasing both the magnitude and duration of oxygen decrease. Therefore, under the influence of alcohol, the use of opioid drugs becomes much more dangerous, increasing brain hypoxia and enhancing the probability of serious health complications, including coma and death.


Brain Chemistry/drug effects , Ethanol/pharmacology , Heroin/toxicity , Hypoxia/chemically induced , Narcotics/toxicity , Oxygen Consumption/drug effects , Administration, Intravenous , Animals , Dose-Response Relationship, Drug , Drug Interactions , Heroin/administration & dosage , Hypoxia/metabolism , Male , Narcotics/administration & dosage , Rats , Rats, Long-Evans , Substance Abuse, Intravenous
12.
Brain Res Bull ; 174: 230-239, 2021 09.
Article En | MEDLINE | ID: mdl-34175385

Increased microglial NADPH oxidase (NOX2) production may make an important contribution to the increased incidence and severity of ischemic stroke associated with diabetes. Imidazoline receptors are closely associated with neuroprotection, but the neuroprotective effects of the selective I2-imidazoline receptor ligand 2-(2-benzofuranyl)-2-imidazoline (2BFI) in diabetes has not been established. The effect of 2BFI on microglial NOX2 production was investigated using a co-culture of neurons and microglia, and the effect on cerebral ischemia-reperfusion (IR) injury was determined in diabetic rats. Garcia neurological scores, brain infarct volumes, brain water content, TUNEL staining, blood-brain barrier, and immunofluorescent labeling for microglia were evaluated. Western blots were used to determine gp91phox and Tyr1472 expression. Anti-inflammatory cytokine (IL-10) and inflammatory cytokine secretion was determined using ELISA kits. The brain infarct volumes, TUNEL-positive neurons, expression of microglia, brain water content, blood-brain barrier structure damage, and gp91phox and Tyr1472 expression were increased, the Garcia neurological scores were significantly decreased in the IR group, and 2BFI relieved these alterations. The IL-10 concentration was increased in the IR group; 2BFI significantly improved this increase. The neuron apoptosis and necrosis rates, and production of reactive oxygen species (ROS) and inflammatory cytokines, including IL-6, IL-8, TNF-α, and 8-iso-PGF2α, were significantly increased by high glucose stimulation combined with oxygen-glucose deprivation treatment, which were inhibited by 2BFI. The 2BFI ameliorated cerebral ischemia-reperfusion injury in diabetes and decreased neuron death in an in vitro model. The mechanism underlying these findings may be related to the decreased production of inflammatory factors and reactive oxygen species from microglia.


Benzofurans/therapeutic use , Diabetes Complications/prevention & control , Imidazoles/therapeutic use , Microglia/drug effects , Neuroprotective Agents/therapeutic use , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Blood-Brain Barrier/pathology , Body Water/metabolism , Brain Chemistry/drug effects , Brain Infarction/pathology , Coculture Techniques , Cytokines/metabolism , Diabetes Mellitus, Experimental/complications , Male , NADPH Oxidase 2/metabolism , Necrosis , Neurons/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reperfusion Injury/complications
13.
Indian J Pharmacol ; 53(2): 132-142, 2021.
Article En | MEDLINE | ID: mdl-34100397

OBJECTIVES: Water contaminated with arsenic affected millions of people worldwide and arsenic exposure is related to various neurological disorders. Hence, the current study was planned to investigate the neuroprotective activity of diosmin (DSN) against arsenic induced neurotoxicity as an attempt to identify therapeutic intervention to combat arsenicism. MATERIALS AND METHODS: Sodium arsenite an inducer of neurotoxicity was administered orally (13 mg/kg) and DSN treatment at two selected doses (50 and 100 mg/kg) was done for 21 days. Behavioral and biochemical variations were examined by various parameters. Furthermore, histopathological and immunohistochemistry studies were done with the brain sections. RESULTS: The behavioral studies evidenced that arsenic has suppressed the exploratory behavior and motor coordination in rats and DSN treatment has recovered the behavioral changes to normal. Arsenic administration has also found to induce oxidative stress and DSN co-treatment has ameliorated the oxidative stress markers. Interestingly, depleted levels of neurotransmitters were observed with the arsenic and it was restored back by the DSN treatment. Histopathological alterations like pyknosis of the neuronal cells were identified with arsenic exposure and subsided upon DSN co administration. Immunohistochemical studies have revealed the expression of NOX4 and its gp91phox and P47phox subunits and its suppression by DSN treatment may be the key therapeutic factor of it. CONCLUSIONS: Treatment with DSN showed a beneficial effect in protecting against arsenic-induced neurotoxicity by suppressing the toxicity changes and the antioxidant effect of DSN might be attributed to its ability of suppressing NOX4 and its subunits.


Arsenic/toxicity , Diosmin/therapeutic use , NADPH Oxidase 4/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Animals , Antioxidants/analysis , Arsenic/analysis , Brain/drug effects , Brain/pathology , Brain Chemistry/drug effects , Female , Maze Learning/drug effects , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology , Neurotransmitter Agents/analysis , Oxidative Stress/drug effects , Protein Subunits/antagonists & inhibitors , Rats , Rats, Wistar
14.
J Neurochem ; 158(4): 912-927, 2021 08.
Article En | MEDLINE | ID: mdl-34050937

Post-traumatic stress disorder (PTSD) is characterized by depression/anxiety and memory failure, primarily fear memory. According to the reports, neuroinflammation and synaptic plasticity can play a role in the neurophysiological mechanisms underlying PTSD. Bromodomain-containing protein 4 (Brd4) intriguingly affects regulating of inflammatory responses and learning and memory. This study aimed to explore the effect of inhibiting Brd4 on depression/anxiety-like behaviors, spatial and fear memory, and underlying mechanisms in a model of PTSD. Inescapable foot shocks (IFS) with a sound reminder in 6 days were used to induce PTSD-like behaviors which were tested using contextual and cue fear tests, sucrose preference test, open-field test, elevated plus maze test, and Y-maze test. Meanwhile, the Brd4 inhibitor JQ1 was used as an intervention. The results found that IFS induced PTSD-like behaviors and indicated obvious Brd4 expression in microglia of the prefrontal cortex (PFC), hippocampus, and amygdala, pro-inflammatory cytokines over-expression, microglial activation, and nuclear factor-kappa B over-expression in PFC and hippocampus but not in amygdala. Meanwhile, the alterations of immediate early genes (IEGs) were found in PFC, hippocampus, and amygdala. Besides, dendritic spine density was reduced in PFC and hippocampus but was elevated in amygdala of rats with IFS. In addition, treatment with JQ1 significantly reduced freezing time in the contextual and cue fear test, reversed the behavioral impairment, decreased the elevated neuroinflammation, and normalized the alteration in IEGs and dendritic spine densities. The results suggested that Brd4 was involved in IFS-induced PTSD-like behaviors through regulating neuroinflammation, dynamics of IEGs, and synaptic plasticity.


Encephalitis/drug therapy , Fear/psychology , Gene Expression Regulation/drug effects , Genes, Immediate-Early/drug effects , Nuclear Proteins/antagonists & inhibitors , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Transcription Factors/antagonists & inhibitors , Animals , Anxiety/drug therapy , Anxiety/psychology , Azepines/pharmacology , Azepines/therapeutic use , Brain Chemistry/drug effects , Cues , Dendritic Spines/drug effects , Depression/drug therapy , Depression/psychology , Encephalitis/genetics , Male , Memory/drug effects , Motor Activity/drug effects , Rats , Rats, Wistar , Triazoles/pharmacology , Triazoles/therapeutic use
15.
Brain Res Bull ; 174: 11-21, 2021 09.
Article En | MEDLINE | ID: mdl-33991606

Dexmedetomidine (Dex) has been suggested to exert a protective function in ischemic brain injury. In this study, we aimed to elucidate the intrinsic mechanisms of Dex in regulating microglia pyroptosis in ischemic brain injury via the purinergic 2X7 receptor (P2X7R)/NLRP3/Caspase-1 signaling pathway. First, permanent middle cerebral artery occlusion (p-MCAO) rat model was established, followed by the measurement of behavioral deficit, neuronal injury, the volume of brain edema and the infarct size. Dex treatment was suggested to alleviate the neurological deficits in p-MCAO rats and reduce the brain water content and infarct size. Additionally, rat microglia were cultured in vitro and a model of oxygen and glucose (OGD) was established. Microglia cell activity and ultrastructure were detected. Dex could increase cell activity and reduce LDH activity, partially reversing the changes in cell morphology. Furthermore, the activation of P2X7R/NLRP3/Caspase-1 pathway was tested. The obtained findings indicated Dex suppressed microglial pyroptosis by inhibiting the P2X7R/NLRP3/Caspase-1 pathway. Inhibition of P2X7R or NLRP3 could inhibit Caspase-1 p10 expression, improve cell activity, and reduce LDH activity. The same result was verified in vivo experiments. This study indicated that Dex inhibited microglia pyroptosis by blocking the P2X7R/NLRP3/Caspase-1 pathway, thus playing a protective role against ischemic brain injury.


Adrenergic alpha-2 Receptor Agonists/pharmacology , Brain Ischemia/prevention & control , Caspase 1/drug effects , Dexmedetomidine/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Neuroprotective Agents/pharmacology , Receptors, Purinergic P2X7/drug effects , Signal Transduction/drug effects , Animals , Brain Chemistry/drug effects , Brain Edema/prevention & control , Brain Ischemia/psychology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/psychology , Male , Microglia/metabolism , Microglia/pathology , Pyroptosis/drug effects , Rats, Sprague-Dawley
16.
Psychopharmacology (Berl) ; 238(8): 2349-2364, 2021 Aug.
Article En | MEDLINE | ID: mdl-34032876

RATIONALE: 4-Iodo-2,5-dimethoxy-N-(2-methoxybenzyl)phenethylamine (25I-NBOMe) is a potent serotonin 5-HT2A/2C receptor agonist with hallucinogenic activity. There is no data on the 25I-NBOMe effect on brain neurotransmission and animal performance after chronic administration. OBJECTIVES: We examined the effect of a 7-day treatment with 25I-NBOMe (0.3 mg/kg/day) on neurotransmitters' release and rats' behavior in comparison to acute dose. METHODS: Changes in dopamine (DA), serotonin (5-HT), acetylcholine (ACh), and glutamate release were studied using microdialysis in freely moving rats. The hallucinogenic activity was measured in the wet dog shake (WDS) test. The animal locomotion was examined in the open field (OF) test, short-term memory in the novel object recognition (NOR) test. The anxiogenic/anxiolytic properties of the drug were tested using the light/dark box (LDB) test. RESULTS: Repeated administration of 25I-NBOMe decreased the response to a challenge dose of DA, 5-HT, and glutamatergic neurons in the frontal cortex as well as weakened the hallucinogenic activity in comparison to acute dose. In contrast, striatal and accumbal DA and 5-HT release and accumbal but not striatal glutamate release in response to the challenge dose of 25I-NBOMe was increased in comparison to acute treatment. The ACh release was increased in all brain regions. Behavioral tests showed a motor activity reduction and memory deficiency in comparison to a single dose and induction of anxiety after the drug's chronic and acute administration. CONCLUSIONS: Our findings suggest that multiple injections of 25I-NBOMe induce tolerance to hallucinogenic activity and produce alterations in neurotransmission. 25I-NBOMe effect on short-term memory, locomotor function, and anxiety seems to be the result of complex interactions between neurotransmitter pathways.


Brain Chemistry/drug effects , Dimethoxyphenylethylamine/analogs & derivatives , Hallucinogens/pharmacology , Locomotion/drug effects , Animals , Brain Chemistry/physiology , Dimethoxyphenylethylamine/pharmacology , Dopamine/metabolism , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Glutamic Acid/metabolism , Locomotion/physiology , Male , Microdialysis/methods , Rats , Rats, Wistar , Serotonin/metabolism
17.
Brain Res ; 1764: 147480, 2021 08 01.
Article En | MEDLINE | ID: mdl-33861997

It has been suggested that cannabis consumption during adolescence may be an initial step to cocaine use in adulthood. Indeed, previous preclinical data show that adolescent exposure to cannabinoids (both natural and synthetic) potentiates cocaine self-administration in rats. Here we aimed at gaining a deeper understanding of the cellular activation patterns induced by cocaine as revealed by Fos imaging and how these patterns may change due to adolescent exposure to THC. Male and female Wistar rats were administered every other day THC (3 mg/kg i.p.) or vehicle from postnatal day 28-44. At adulthood (PND90) they were given an injection of cocaine (20 mg/kg i.p.) or saline and sacrificed 90 min later. Cocaine-induced Fos activation was measured by immunohistochemistry as an index of cellular activation. We found that cocaine-induced activation in the motor cortex was stronger in THC-exposed rats. Moreover, there was significant sex-dependent interaction between cocaine and adolescent THC exposure in the dorsal hypothalamus, suggesting that cocaine induced a more robust cellular activation in THC-exposed females but not in THC-treated males. Other THC- and cocaine-induced effects were also evident. These results add to the previous literature suggesting that the behavioral, cellular, molecular, and brain-activating actions of cocaine are modulated by early experience with cannabinoids and provide additional knowledge that may explain the enhanced actions of cocaine in rats exposed to cannabinoids during their adolescence.


Brain Chemistry/drug effects , Cocaine/pharmacology , Dronabinol/pharmacology , Genes, fos/drug effects , Hallucinogens/pharmacology , Aging , Animals , Behavior, Animal/drug effects , Drug Interactions , Female , Hypothalamus, Posterior/drug effects , Immunohistochemistry , Male , Motor Cortex/drug effects , Motor Cortex/metabolism , Rats , Rats, Wistar , Sex Characteristics
18.
J Ethnopharmacol ; 275: 114142, 2021 Jul 15.
Article En | MEDLINE | ID: mdl-33910044

ETHNOPHARMACOLOGICAL RELEVANCE: Cymbopogon citratus (DC.) Stapf (C. citratus) is consumed as an infusion in folk medicine due to its pharmacological properties and action in the central nervous system. Epilepsy is a neurological disorder that affects millions of people. Since the currently available antiepileptic drugs often cause undesirable side effects, new alternative therapeutic strategies based on medicinal plants have been proposed. AIM OF THE STUDY: This study aimed to investigate the anticonvulsant and neuroprotective effects of C. citratus essential oil (EO) and hydroalcoholic extract (E1) from its leaves, as well as of its related compounds citral (CIT) and geraniol (GER) against the effects of pentylenetetrazole (PTZ) induced seizures in zebrafish (Danio rerio). MATERIALS AND METHODS: To evaluate the anticonvulsant properties of the samples, adult animals were pre-treated (by immersion) and subsequently exposed to PTZ solution. The involvement of GABAA receptors in the antiepileptic effects was investigated by the coadministration of flumazenil (FMZ), a known GABAA receptor antagonist. Oxidative stress markers malondialdehyde (MDA), glutathione (GSH), catalase (CAT) and nitric oxide (NO) were assessed in zebrafish brain homogenates after PTZ exposure. RESULTS: All samples increased the latency time for the first seizure, which was reduced when animals were pretreated with FMZ, suggesting the involvement of GABAA receptors in the observed properties. The association between CIT and GER at the lowest concentration studied showed a synergistic effect on the anticonvulsant activity. Decreases in MDA and NO levels and increases in GSH and CAT levels in the brain of treated animals suggested the neuroprotective effect of the compounds investigated. CONCLUSIONS: Our results proved that C. citratus EO, E1, CIT and GER have anticonvulsant effects in zebrafish and could be used as a promising adjuvant therapeutic strategy for epilepsy treatment. Furthermore, zebrafish demonstrated to be an alternative animal model of epilepsy to evaluate the anticonvulsant and neuroprotective effects of C. citratus.


Acyclic Monoterpenes/pharmacology , Anticonvulsants/pharmacology , Cymbopogon/chemistry , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Seizures/drug therapy , Acyclic Monoterpenes/therapeutic use , Animals , Anticonvulsants/therapeutic use , Brain Chemistry/drug effects , Catalase/metabolism , Disease Models, Animal , Flumazenil/pharmacology , Flumazenil/therapeutic use , Glutathione/metabolism , Malondialdehyde/metabolism , Medicine, Traditional , Neuroprotective Agents/therapeutic use , Oils, Volatile/pharmacology , Oils, Volatile/therapeutic use , Oxidative Stress/drug effects , Pentylenetetrazole/toxicity , Plant Extracts/therapeutic use , Plant Leaves , Receptors, GABA-A/metabolism , Seizures/chemically induced , Zebrafish
19.
Neurotoxicol Teratol ; 86: 106982, 2021.
Article En | MEDLINE | ID: mdl-33845156

Despite reports that quinoline antimalarials including chloroquine (Chq) exhibit idiosyncratic neuropsychiatric effects even at low doses, the drug continues to be in widespread use during pregnancy. Surprisingly, very few studies have examined the potential neurotoxic action of Chq exposure at different points of gestation or how this phenomenon may affect neurophysiological well-being in later life. We therefore studied behavior, and the expression of specific genes and neurochemicals modulating crucial neural processes in offspring of rats exposed to prophylactic dose of Chq during different stages of gestation. Pregnant rats were injected 5 mg/kg/day (3 times) of Chq either during early- (first week), mid- (second week), late- (third week), or throughout- (all weeks) gestation, while controls received PBS injection. Behavioral characterization of offspring between postnatal days 15-20 in the open field, Y-maze, elevated plus and elevated zero mazes revealed that Chq evoked anxiogenic responses and perturbed spatial memory in rats, although locomotor activity was generally unaltered. In the prefrontal cortex (PFC), hippocampus and cerebellum of rats prenatally exposed to Chq, RT-qPCR analysis revealed decreased mRNA expression of presynaptic marker synaptophysin, which was accompanied by downregulation of postsynaptic marker PSD95. Synaptic marker PICK1 expression was also downregulated in the hippocampus but was unperturbed in the PFC and cerebellum. In addition to recorded SOD downregulation in cortical and hippocampal lysates, induction of oxidative stress in rats prenatally exposed to Chq was corroborated by lipid peroxidation as evinced by increased MDA levels. Offspring of rats infused with Chq at mid-gestation and weekly treatment throughout gestation were particularly susceptible to neurotoxic changes, especially in the hippocampus. Interestingly, Chq did not cause histopathological changes in any of the brain areas. Taken together, our findings causally link intrauterine exposure to Chq with postnatal behavioral impairment and neurotoxic changes in rats.


Behavior, Animal/drug effects , Brain Chemistry/drug effects , Chloroquine/toxicity , Neuronal Plasticity/drug effects , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/psychology , Animals , Anxiety/chemically induced , Anxiety/psychology , Female , Gene Expression/drug effects , Gestational Age , Maze Learning/drug effects , Motor Activity/drug effects , Pregnancy , Rats , Spatial Memory/drug effects
20.
Neurotoxicology ; 84: 136-145, 2021 05.
Article En | MEDLINE | ID: mdl-33774067

Mercury is a neurotoxic chemical that represents one of the greatest pollution threats to Arctic ecosystem health. Evaluating the direct neurotoxic effects of mercury in free ranging wildlife is challenging, necessitating the use of neurochemical biomarkers to assess potential sub-clinical neurological changes. The objective of this study was to characterize the distribution and speciation of mercury, as well as exposure-associated changes in neurochemistry, across multiple brain regions (n = 10) and marine mammal species (n = 5) that each occupy a trophic niche in the Arctic ecosystem. We found consistent species differences in mean brain and brain region-specific concentrations of total mercury (THg) and methyl mercury (MeHg), with higher concentrations in toothed whales (narwhal, pilot whales and harbour porpoise) compared to fur-bearing mammals (polar bear and ringed seal). Mean THg (µg/g dw) in decreasing rank order was: pilot whale (11.9) > narwhal (7.7) > harbour porpoise (3.6) > polar bear (0.6) > ringed seal (0.2). The higher THg concentrations in toothed whales was associated with a marked reduction in the percentage of MeHg (<40 %) compared to polar bears (>70 %) that had lower brain THg concentrations. This pattern in mercury concentration and speciation corresponded broadly to an overall higher number of mercury-associated neurochemical biomarker correlations in toothed whales. Of the 226 correlations between mercury and neurochemical biomarkers across brain regions, we found 60 (27 %) meaningful relationships (r>0.60 or p < 0.10). We add to the growing weight of evidence that wildlife accumulate mercury in their brains and demonstrate that there is variance in accumulation across species as well as across distinct brain regions, and that some of these exposures may be associated with sub-clinical changes in neurochemistry.


Brain Chemistry/drug effects , Brain/drug effects , Mercury/analysis , Water Pollutants, Chemical/analysis , Animals , Arctic Regions/epidemiology , Biomarkers/analysis , Biomarkers/chemistry , Brain/physiology , Brain Chemistry/physiology , Mercury/adverse effects , Phocoena , Seals, Earless , Species Specificity , Ursidae , Water Pollutants, Chemical/adverse effects , Whales , Whales, Pilot
...