Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 451: 114526, 2023 08 05.
Article in English | MEDLINE | ID: mdl-37271313

ABSTRACT

This study aimed to evaluate the behavioral and energy metabolism parameters in female mice subjected to obesity and offspring deprivation (OD) stress. Eighty female Swiss mice, 40 days old, were weighed and divided into two groups: Control group (control diet, n = 40) and Obese group (high-fat diet, n = 40), for induction of the animal model of obesity, the protocol was based on the consumption of a high-fat diet and lasted 8 weeks. Subsequently, the females were subjected to pregnancy, after the birth of the offspring, were divided again into the following groups (n = 20): Control non-deprived (ND), Control + OD, Obese ND, and Obese + OD, for induction of the stress protocol by OD. After the offspring were 21 days old, weaning was performed and the dams were subjected to behavioral tests. The animals were humanely sacrificed, the brain was removed, and brain structures were isolated to assess energy metabolism. Both obesity and OD led to anhedonia in the dams. It was shown that the structures most affected by obesity and OD are the hypothalamus and hippocampus, as evidenced by the mitochondrial dysfunction found in these structures. When analyzing the groups separately, it was observed that OD led to more pronounced mitochondrial damage; however, the association of obesity with OD, as well as obesity alone, also generated damage. Thus, it is concluded that obesity and OD lead to anhedonia in animals and to mitochondrial dysfunction in the hypothalamus and hippocampus, which may lead to losses in feeding control and cognition of the dams.


Subject(s)
Anhedonia , Prenatal Exposure Delayed Effects , Pregnancy , Mice , Female , Animals , Humans , Obesity/metabolism , Diet, High-Fat/adverse effects , Weaning , Energy Metabolism
2.
Behav Brain Res ; 434: 114019, 2022 09 26.
Article in English | MEDLINE | ID: mdl-35872330

ABSTRACT

Obesity is associated with low-grade chronic inflammation and oxidative stress, affecting the brain's reward system by decreasing dopaminergic neurotransmission. It is known that dopaminergic neurotransmission is also reduced in Parkinson's disease (PD), and high adiposity is considered a risk factor for the development of several neurodegenerative diseases, including PD. This study aimed to assess the effects of obesity on neuroinflammatory and neurochemical parameters in an animal model of reserpine-induced PD. The obese group showed increased inflammation and oxidative damage as well as inhibition of mitochondrial respiratory chain complexes I and II and DNA damage in the evaluated structures. The PD group did not show inflammation or mitochondrial dysfunction but exhibited oxidative damage in the hippocampus. The combination group (obesity + PD) showed reduced inflammation and oxidative stress and increased activity of complexes I and II of the mitochondrial respiratory chain in most of the analyzed structures. On the other hand, obesity + PD caused oxidative damage to proteins in the liver, prefrontal cortex, striatum, and cerebral cortex and oxidative stress in the hypothalamus, resulting in reduced catalase activity. Furthermore, the combination group showed DNA damage in blood, liver, and cerebral cortex. In conclusion, it was observed that the association of obesity and PD did not increase inflammation, oxidative stress, or mitochondrial dysfunction in most of the evaluated structures but increased oxidative damage and induced mechanisms that led to DNA damage in peripheral tissues and brain structures.


Subject(s)
Parkinson Disease , Animals , Disease Models, Animal , Inflammation , Obesity , Oxidative Stress , Reserpine
3.
Front Psychiatry ; 12: 638032, 2021.
Article in English | MEDLINE | ID: mdl-33643100

ABSTRACT

Although cannabis has been known for ages as an "alternative medicine" to provide relief from seizures, pain, anxiety, and inflammation, there had always been a limited scientific review to prove and establish its use in clinics. Early studies carried out by Carlini's group in Brazil suggested that cannabidiol (CBD), a non-psychotropic phytocannabinoid present in Cannabis sativa, has anticonvulsant properties in animal models and reduced seizure frequency in limited human trials. Over the past few years, the potential use of cannabis extract in refractory epilepsy, including childhood epilepsies such as Dravet's syndrome and Lennox-Gastaut Syndrome, has opened a new era of treating epileptic patients. Thus, a considerable number of pre-clinical and clinical studies have provided strong evidence that phytocannabinoids has anticonvulsant properties, as well as being promising in the treatment of different neuropsychiatric disorders, such as depression, anxiety, post-traumatic stress disorder (PTSD), addiction, neurodegenerative disorders and autism spectrum disorder (ASD). Given the advances of cannabinoids, especially CBD, in the treatment of epilepsy, would the same expectation regarding the treatment of other neuropsychiatric disorders be possible? The present review highlights some contributions from Brazilian researchers and other studies reported elsewhere on the history, pre-clinical and clinical data underlying the use of cannabinoids for the already widespread treatment of refractory epilepsies and the possibility of use in the treatment of some neuropsychiatric disorders.

4.
An Acad Bras Cienc ; 92(4): e20200837, 2020.
Article in English | MEDLINE | ID: mdl-33295581

ABSTRACT

The intrauterine environment is infl uenced by several factors, genetic or environmental, which are essential in understanding the pathophysiological mechanisms of some diseases. In this study, the aim was to investigate the impact of prenatal lipopolysaccharide exposure on the development of rats. Fifty pregnant rats received intraperitoneal administration of lipopolysaccharide (100 µg/kg), or saline at the same dose, on the 9.5th day of pregnancy. The offspring of these rats were analyzed for indicators of brain and somatic development and maturation of physical characteristics. Refl ex ontogenesis was also analyzed by vibrissae placement, negative geotaxis, palmar grasp, precipice aversion, decubitus recovery and acceleration reaction. Administration of lipopolysaccharide on the 9.5th gestational day caused delayed opening of the auditory pavilion, reduction in the length of the tail, body, cranial axes, and body weight. Thus, maternal infections can interfere in the intrauterine environment, impairing functional and structural aspects of the central nervous system, as well as the maturation of physical characteristics.


Subject(s)
Lipopolysaccharides , Prenatal Exposure Delayed Effects , Age Factors , Animals , Animals, Newborn , Behavior, Animal , Body Weight , Female , Lipopolysaccharides/toxicity , Pregnancy , Rats , Tail
5.
Front Neurosci ; 12: 502, 2018.
Article in English | MEDLINE | ID: mdl-30087591

ABSTRACT

Post-traumatic stress disorder (PTSD) is characterized by poor adaptation to a traumatic experience. This disorder affects approximately 10% of people at some point in life. Current pharmacological therapies for PTSD have been shown to be inefficient and produce considerable side effects. Since the discovery of the involvement of the endocannabinoid (eCB) system in emotional memory processing, pharmacological manipulation of eCB signaling has become a therapeutic possibility for the treatment of PTSD. Cannabidiol (CBD), a phytocannabinoid constituent of Cannabis sativa without the psychoactive effects of Δ9-tetrahydrocannabinol, has gained particular attention. Preclinical studies in different rodent behavioral models have shown that CBD can both facilitate the extinction of aversive memories and block their reconsolidation, possibly through potentialization of the eCB system. These results, combined with the currently available pharmacological treatments for PTSD being limited, necessitated testing CBD use with the same therapeutic purpose in humans as well. Indeed, as observed in rodents, recent studies have confirmed the ability of CBD to alter important aspects of aversive memories in humans and promote significant improvements in the symptomatology of PTSD. The goal of this review was to highlight the potential of CBD as a treatment for disorders related to inappropriate retention of aversive memories, by assessing evidence from preclinical to human experimental studies.

6.
Behav Brain Res ; 316: 74-81, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27566183

ABSTRACT

Previous studies have demonstrated that targeting bradykinin receptors is a promising strategy to counteract the cognitive impairment related with aging and Alzheimer's disease (AD). The hippocampus is critical for cognition, and abnormalities in this brain region are linked to the decline in mental ability. Nevertheless, the impact of bradykinin signaling on hippocampal function is unknown. Therefore, we sought to determine the role of hippocampal bradykinin receptors B1R and B2R on the cognitive decline of middle-aged rats. Twelve-month-old rats exhibited impaired ability to acquire and retrieve spatial information in the Morris water maze task. A single intra-hippocampal injection of the selective B1R antagonist des-Arg9-[Leu8]-bradykinin (DALBK, 3 nmol), but not the selective B2R antagonist D-Arg-[Hyp3,Thi5,D-Tic7,Oic8]-BK (Hoe 140, 3 nmol), reversed the spatial learning and memory deficits on these animals. However, both drugs did not affect the cognitive function in 3-month-old rats, suggesting absence of nootropic properties. Molecular biology analysis revealed an up-regulation of B1R expression in the hippocampal CA1 sub-region and in the pre-frontal cortex of 12-month-old rats, whereas no changes in the B2R expression were observed in middle-aged rats. These findings provide new evidence that inappropriate hippocampal B1R expression and activation exert a critical role on the spatial learning and memory deficits in middle-aged rats. Therefore, selective B1R antagonists, especially orally active non-peptide antagonists, may represent drugs of potential interest to counteract the age-related cognitive decline.


Subject(s)
Hippocampus/metabolism , Memory Disorders/pathology , Receptor, Bradykinin B1/metabolism , Spatial Learning/drug effects , Aging/drug effects , Animals , Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Bradykinin B1 Receptor Antagonists/pharmacology , Disease Models, Animal , Hippocampus/drug effects , Male , Maze Learning/drug effects , Memory Disorders/drug therapy , Rats , Rats, Wistar , Up-Regulation/drug effects
7.
Neurochem Int ; 90: 72-84, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26196379

ABSTRACT

Corticosteroid and endocannabinoid actions converge on prefrontocortical circuits associated with neuropsychiatric illnesses. Corticosteroids can also modulate forebrain synapses by using endocannabinoid effector systems. Here, we determined whether corticosteroids can modulate transmitter release directly in the frontal cortex and, in doing so, whether they affect presynaptic CB1 cannabinoid receptor- (CB1R) mediated neuromodulation. By Western blotting of purified subcellular fractions of the rat frontal cortex, we found glucocorticoid receptors (GcRs) and CB1Rs enriched in isolated frontocortical nerve terminals (synaptosomes). CB1Rs were predominantly presynaptically located while GcRs showed preference for the post-synaptic fraction. Additional confocal microscopy analysis of cortical and hippocampal regions revealed vesicular GABA transporter-positive and vesicular glutamate transporter 1-positive nerve terminals endowed with CB1R immunoreactivity, apposing GcR-positive post-synaptic compartments. In functional transmitter release assay, corticosteroids, corticosterone (0.1-10 microM) and dexamethasone (0.1-10 microM) did not significantly affect the evoked release of [(3)H]GABA and [(14)C]glutamate in superfused synaptosomes, isolated from both rats and mice. In contrast, the synthetic cannabinoid, WIN55212-2 (1 microM) diminished the release of both [(3)H]GABA and [(14)C]glutamate, evoked with various depolarization paradigms. This effect of WIN55212-2 was abolished by the CB1R neutral antagonist, O-2050 (1 microM), and was absent in the CB1R KO mice. CB2R-selective agonists did not affect the release of either neurotransmitter. The lack of robust presynaptic neuromodulation by corticosteroids was unchanged upon either CB1R activation or genetic inactivation. Altogether, corticosteroids are unlikely to exert direct non-genomic presynaptic neuromodulation in the frontal cortex, but they may do so indirectly, via the stimulation of trans-synaptic endocannabinoid signaling.


Subject(s)
Benzoxazines/pharmacology , Frontal Lobe/drug effects , Morpholines/pharmacology , Naphthalenes/pharmacology , Receptor, Cannabinoid, CB1/drug effects , Synapses/drug effects , Animals , Endocannabinoids/metabolism , Frontal Lobe/metabolism , Glutamic Acid/metabolism , Male , Mice , Presynaptic Terminals/metabolism , Rats, Wistar , Receptor, Cannabinoid, CB1/deficiency , Receptor, Cannabinoid, CB1/metabolism , Receptors, Presynaptic/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism
8.
Behav Brain Res ; 271: 325-32, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24975422

ABSTRACT

Chronic consumption of drugs with addictive potential induces profound synaptic changes in the dopaminergic mesocorticolimbic pathway that underlie the long-term behavioral alterations seen in addicted subjects. Thus, exploring modulation systems of dopaminergic function may reveal novel targets to interfere with drug addiction. We recently showed that cellular prion protein (PrP(C)) affects the homeostasis of the dopaminergic system by interfering with dopamine synthesis, content, receptor density and signaling pathways in different brain areas. Here we report that the genetic deletion of PrP(C) modulates ethanol (EtOH)-induced behavioral alterations including the maintenance of drug seeking, voluntary consumption and the development of EtOH tolerance, all pivotal steps in drug addiction. Notably, these behavioral changes were accompanied by a significant depletion of dopamine levels in the prefrontal cortex and reduced dopamine D1 receptors in PrP(C) knockout mice. Furthermore, the pharmacological blockade of dopamine D1 receptors, but not D2 receptors, attenuated the abnormal EtOH consumption in PrP(C) knockout mice. Altogether, these findings provide new evidence that the PrP(C)/dopamine interaction plays a pivotal role in EtOH addictive properties in mice.


Subject(s)
Adaptation, Psychological/drug effects , Alcohol Drinking/psychology , Dopamine/deficiency , Ethanol/pharmacology , PrPC Proteins/deficiency , Alcohol Drinking/genetics , Animals , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Female , Male , Mice , Mice, Knockout , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism
9.
Behav Brain Res ; 250: 23-7, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23643693

ABSTRACT

Previous studies have implicated cannabinoids in extinction of conditioned fear. We have recently showed that intraventricular infusion of the phytocannabinoid cannabidiol (CBD) facilitates fear extinction, but the brain regions underlying this effect remained unknown. Here we demonstrate that repeated microinjections of CBD into the infralimbic cortex (IL) facilitated fear extinction, as indicated by reduced levels of freezing during extinction test. Systemic administration of the CB1 receptor antagonist rimonabant blocked the effects of intra-IL CBD, suggesting that CBD acts through CB1 receptors to facilitate fear extinction. Our findings suggest a potential therapeutic use of CBD for extinction-based therapies of aversive memories in humans.


Subject(s)
Cannabidiol/pharmacology , Cerebral Cortex/drug effects , Conditioning, Classical/drug effects , Extinction, Psychological/drug effects , Fear/drug effects , Receptor, Cannabinoid, CB1/metabolism , Animals , Cannabinoid Receptor Antagonists/pharmacology , Cerebral Cortex/physiology , Dose-Response Relationship, Drug , Drug Interactions , Electroshock/adverse effects , Fear/psychology , Freezing Reaction, Cataleptic/drug effects , Male , Microinjections , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Long-Evans , Rimonabant , Time Factors
10.
Eur Neuropsychopharmacol ; 18(12): 849-59, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18706790

ABSTRACT

The present study investigated the central effects of the eCB uptake/metabolism inhibitor AM404 and the phytocannabinoid cannabidiol (CBD) on the extinction of contextual fear memories in rats. Rats were conditioned and 24 h later subjected to three consecutive 9-min non-reinforced exposures to the conditioning context (extinction sessions, 24 h intervals). AM404 or CBD was injected i.c.v. 5 min before each extinction session and a 3-min drug-free test of contextual memory was performed 24 h after the last extinction session. AM404 (1.0 microg/microl, i.c.v.) and CBD (2.0 microg/microl, i.c.v.) facilitated extinction of contextual fear memory, with persistent effects. These responses were antagonized by the CB1-selective antagonist SR141716A (0.2 mg/kg, i.p.), but not by the TRPV1-selective antagonist capsazepine (5.0 microg/microl, i.c.v.). The effect of the anxiolytic drug Diazepam (DZP) on the extinction of contextual fear memory was also investigated. In contrast with the CBD and AM404 results, DZP induced a general reduction in the expression of conditioned freezing. Both AM404 and CBD induced anti-anxiogenic effect in the fear-potentiated plus-maze test, whereas DZP was anxiolytic in conditioned and unconditioned rats. In conclusion, CBD, a non-psychoactive phytocannabinoid could be an interesting pharmacological approach to reduce the anxiogenic effects of stress and promote the extinction of fear memories.


Subject(s)
Anti-Anxiety Agents/pharmacology , Arachidonic Acids/pharmacology , Cannabidiol/pharmacology , Conditioning, Psychological/drug effects , Extinction, Psychological/drug effects , Fear , Analysis of Variance , Animals , Behavior, Animal/drug effects , Cannabidiol/antagonists & inhibitors , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Freezing Reaction, Cataleptic/drug effects , Injections, Intraventricular/methods , Male , Maze Learning/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Rimonabant , TRPV Cation Channels/antagonists & inhibitors
11.
Neurobiol Learn Mem ; 90(1): 290-3, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18486502

ABSTRACT

Facilitation of memory extinction by manipulation of the endocannabinoid (eCB) system has been recently studied in several paradigms. Our previous results pointed to facilitation of contextual fear memory extinction by a low dose of a cannabinoid agonist, with a suggestion of short-term effects. The aim of the present study was to further investigate the effects of cannabinoid drugs in the short- and long-term extinction of conditioned fear using an extended extinction protocol. Male Wistar rats were placed in a conditioning chamber and after 3min received a footshock (1.5mA, 1s). On the next day, they received i.p. drug treatment (WIN55212-2 0.25mg/kg, AM404 10mg/kg, SR141716A 1mg/kg) and were re-exposed to the conditioning chamber for 30min (extinction training). No-Extinction groups received the same drug treatment, but were exposed for 3min to the conditioning chamber. A drug-free test of contextual memory (3min) was performed 7 days later. The cannabinoid agonist WI55212-2 and the inhibitor of eCB metabolism/uptake AM404 facilitated short-term extinction. In addition, long-term effects induced by treatments with WIN55212 and AM404 were completely divergent to those of SR141716A treatment. The present results confirm and extend previous findings showing that the eCB system modulates short-term fear memory extinction with long-lasting consequences.


Subject(s)
Cannabinoids/pharmacology , Extinction, Psychological/drug effects , Fear/physiology , Memory/drug effects , Animals , Arachidonic Acids/pharmacology , Benzoxazines/pharmacology , Calcium Channel Blockers/pharmacology , Cannabinoids/antagonists & inhibitors , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Extinction, Psychological/physiology , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Male , Memory/physiology , Morpholines/pharmacology , Naphthalenes/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Rimonabant , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...