Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Neurosci Lett ; 820: 137589, 2024 Jan 18.
Article En | MEDLINE | ID: mdl-38101612

Depending on its duration and severity, stress may contribute to neuropsychiatric diseases such as depression and anxiety. Studies have shown that stress impacts the hypothalamic-pituitary-adrenal (HPA) axis, but its downstream molecular, behavioral, and nociceptive effects remain unclear. We hypothesized that a 2-hour single exposure to acute restraint stress (ARS) activates the HPA axis and changes DNA methylation, a molecular mechanism involved in the machinery of stress regulation. We further hypothesized that ARS induces anxiety-like and risk assessment behavior and alters nociceptive responses in the rat. We employed biochemical (radioimmunoassay for corticosterone; global DNA methylation by enzyme immunoassay and western blot for DNMT3a expression in the amygdala, ventral hippocampus, and prefrontal cortex) and behavioral (elevated plus maze and dark-light box for anxiety and hot plate test for nociception) tests in adult male Wistar rats exposed to ARS or handling (control). All analyses were performed 24 h after ARS or handling. We found that ARS increased corticosterone levels in the blood, increased the expression of DNMT3a in the prefrontal cortex, promoted anxiety-like and risk assessment behaviors in the elevated plus maze, and increased the nociceptive threshold observed in the hot plate test. Our findings suggest that ARS might be a helpful rat model for studying acute stress and its effects on physiology, epigenetic machinery, and behavior.


Corticosterone , Hypothalamo-Hypophyseal System , Rats , Male , Animals , Hypothalamo-Hypophyseal System/metabolism , Rats, Wistar , Stress, Psychological/psychology , Pituitary-Adrenal System/metabolism , Brain/metabolism , Anxiety/metabolism , Restraint, Physical/psychology
2.
Behav Brain Res ; 458: 114764, 2024 02 26.
Article En | MEDLINE | ID: mdl-37972712

Doxycycline is an antibiotic that has shown neuroprotective, anti-inflammatory, and antidepressant-like effects. Low doses of doxycycline revert the behavioral and neuroinflammatory responses induced by lipopolysaccharide treatment in a mice model of depression. However, the molecular mechanisms involved in the antidepressant action of doxycycline are not yet understood. Doxycycline inhibits the synthesis of nitric oxide (NO), which increases after stress exposure. Inducible NO synthase (iNOS) inhibition also causes antidepressant-like effects in animal models sensitive to antidepressant-like effects such as the forced swimming test (FST). However, no direct study has yet investigated if the antidepressant-like effects of doxycycline could involve changes in NO-mediated neurotransmission. Therefore, this study aimed at investigating: i) the behavioral effects induced by doxycycline alone or in association with ineffective doses of a NO donor (sodium nitroprusside, SNP) or an iNOS inhibitor (1400 W) in mice subjected to the FST; and ii) doxycycline effects in NO metabolite levels in the prefrontal cortex and hippocampus these animals. Male mice (8 weeks) received i.p. injection of saline or doxycycline (10, 30, and 50 mg/kg), alone or combined with SNP (0.1, 0.5, and 1 mg/kg) or 1400 W (1, 3, and 10 µg/kg), and 30 min later were submitted to the FST. Animals were sacrificed immediately after, and NO metabolites nitrate/nitrite (NOx) were measured in the prefrontal cortex and hippocampus. Doxycycline (50 mg/kg) reduced both the immobility time in the FST and NOx levels in the prefrontal cortex of mice compared to the saline group. The antidepressant-like effect of doxycycline in the FST was prevented by SNP (1 mg/kg) pretreatment. Additionally, sub-effective doses of doxycycline (30 mg/kg) associated with 1400 W (1 µg/kg) induced an antidepressant-like effect in the FST. Altogether, our data suggest that the reducing NO levels in the prefrontal cortex through inhibition of iNOS could be related to acute doxycycline treatment resulting in rapid antidepressant-like effects in mice.


Doxycycline , Nitric Oxide , Male , Mice , Animals , Nitric Oxide/metabolism , Doxycycline/pharmacology , Depression/drug therapy , Depression/metabolism , Antidepressive Agents/therapeutic use , Swimming , Prefrontal Cortex/metabolism
3.
Neurochem Int ; 162: 105442, 2023 01.
Article En | MEDLINE | ID: mdl-36402294

S-adenosyl-l-methionine (SAMe), a methyl donor, induces antidepressant effects in preclinical and clinical studies of depression. However, the mechanisms behind these effects have been poorly investigated. Since SAMe is involved in monoamine metabolism, this work aimed at 1) testing the effects induced by systemic treatment with SAMe in mice submitted to the forced swimming test (FST) and tail suspension test (TST); 2) investigating the involvement of serotonergic neurotransmission in the behavioral effects induced by SAMe. To do that, male Swiss mice received systemic injections (1 injection/day, 1 or 7 days) of imipramine (30 mg/kg), L-methionine (400, 800, 1600, and 3200 mg/kg), SAMe (10, 25, 50, 100, and 200 mg/kg), or vehicle (10 ml/kg) and were submitted to the FST or TST, 30 min after the last injection. The effect of SAMe (50 mg/kg) was further investigated in independent groups of male Swiss mice pretreated with p-chlorophenylalanine (PCPA, serotonin synthesis inhibitor, 150 mg/kg daily, 4 days) or with WAY100635 (5-HT1A receptor antagonist, 0.1 mg/kg, 1 injection). One independent group was submitted to the FST and euthanized immediately after for collection of brain samples for neurochemical analyses. Serotonin (5-HT) and noradrenaline (NA) levels were measured in the hippocampus (HPC) and prefrontal cortex (PFC). Furthermore, to investigate if the treatments used could induce any significant exploratory/motor effect which would interfere with the FST results, the animals were also submitted to the open field test (OFT). The administration of imipramine (30 mg/kg), L-methionine (400, 800, 1600, and 3200 mg/kg), and SAMe (10 and 50 mg/kg) reduced the immobility time in the FST, an effect blocked by pretreatment with PCPA and WAY100635. None of the treatments increased the locomotion in the OFT. In conclusion, our results suggest that the antidepressant-like effects induced by SAMe treatment are dependent on serotonin synthesis and 5-HT1A receptor activation.


S-Adenosylmethionine , Serotonin , Male , Mice , Animals , Serotonin/metabolism , S-Adenosylmethionine/pharmacology , Imipramine/pharmacology , Depression/drug therapy , Depression/metabolism , Receptor, Serotonin, 5-HT1A , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Serotonin Antagonists/pharmacology , Selective Serotonin Reuptake Inhibitors , Swimming
4.
Genes (Basel) ; 13(11)2022 11 20.
Article En | MEDLINE | ID: mdl-36421839

Cannabidiol (CBD) is a non-psychotomimetic compound present in cannabis sativa. Many recent studies have indicated that CBD has a promising therapeutic profile for stress-related psychiatric disorders, such as anxiety, schizophrenia and depression. Such a diverse profile has been associated with its complex pharmacology, since CBD can target different neurotransmitter receptors, enzymes, transporters and ion channels. However, the precise contribution of each of those mechanisms for CBD effects is still not yet completely understood. Considering that epigenetic changes make the bridge between gene expression and environment interactions, we review and discuss herein how CBD affects one of the main epigenetic mechanisms associated with the development of stress-related psychiatric disorders: DNA methylation (DNAm). Evidence from in vivo and in silico studies indicate that CBD can regulate the activity of the enzymes responsible for DNAm, due to directly binding to the enzymes and/or by indirectly regulating their activities as a consequence of neurotransmitter-mediated signaling. The implications of this new potential pharmacological target for CBD are discussed in light of its therapeutic and neurodevelopmental effects.


Cannabidiol , Cannabis , Psychiatry , Humans , Cannabidiol/pharmacology , Cannabidiol/therapeutic use , DNA Methylation/genetics , Computer Simulation
5.
Eur J Neurosci ; 55(9-10): 2421-2434, 2022 05.
Article En | MEDLINE | ID: mdl-33170977

It has been postulated that the activation of NMDA receptors (NMDAr) and nitric oxide (NO) production in the hippocampus is involved in the behavioral consequences of stress. Stress triggers NMDAr-induced calcium influx in limbic areas, such as the hippocampus, which in turn activates neuronal NO synthase (nNOS). Inhibition of nNOS or NMDAr activity can prevent stress-induced effects in animal models, but the molecular mechanisms behind this effect are still unclear. In this study, cultured hippocampal neurons treated with NMDA or dexamethasone showed an increased of DNA methyltransferase 3b (DNMT3b) mRNA expression, which was blocked by pre-treatment with nNOS inhibitor nω -propyl-l-arginine (NPA). In rats submitted to the Learned Helplessness paradigm (LH), we observed that inescapable stress increased DNMT3b mRNA expression at 1h and 24h in the hippocampus. The NOS inhibitors 7-NI and aminoguanidine (AMG) decreased the number of escape failures in LH and counteracted the changes in hippocampal DNMT3b mRNA induced in this behavioral paradigm. Altogether, our data suggest that NO produced in response to NMDAr activation following stress upregulates DNMT3b in the hippocampus.


Hippocampus , Nitric Oxide Synthase , Animals , DNA (Cytosine-5-)-Methyltransferases/genetics , Enzyme Inhibitors/pharmacology , Hippocampus/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I , RNA, Messenger/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Physiological , DNA Methyltransferase 3B
6.
Acta Neuropsychiatr ; 33(5): 217-241, 2021 Oct.
Article En | MEDLINE | ID: mdl-34348819

Epigenetic mechanisms such as DNA methylation (DNAm) have been associated with stress responses and increased vulnerability to depression. Abnormal DNAm is observed in stressed animals and depressed individuals. Antidepressant treatment modulates DNAm levels and regulates gene expression in diverse tissues, including the brain and the blood. Therefore, DNAm could be a potential therapeutic target in depression. Here, we reviewed the current knowledge about the involvement of DNAm in the behavioural and molecular changes associated with stress exposure and depression. We also evaluated the possible use of DNAm changes as biomarkers of depression. Finally, we discussed current knowledge limitations and future perspectives.


Biomarkers/blood , Depression/drug therapy , Depression/genetics , Methyltransferases/antagonists & inhibitors , Animals , Antidepressive Agents/pharmacology , Brain/metabolism , CpG Islands , DNA Methylation/drug effects , Depression/blood , Epigenomics , Female , Gene Expression Regulation/drug effects , Humans , Male , Mice , Models, Animal , Rats , Rats, Wistar , Stress, Psychological
7.
Article En | MEDLINE | ID: mdl-34015384

Cannabidiol (CBD), a major non-psychotomimetic component of the Cannabis sativa plant, shows therapeutic potential in several psychiatric disorders, including schizophrenia. The molecular mechanisms underlying the antipsychotic-like effects of CBD are not fully understood. Schizophrenia and antipsychotic treatment can modulate DNA methylation in the blood and brain, resulting in altered expression of diverse genes associated with this complex disorder. However, to date, the possible involvement of DNA methylation in the antipsychotic-like effects of CBD has not been investigated. Therefore, this study aimed at evaluating in mice submitted to the prepulse inhibition (PPI) model: i) the effects of a single injection of CBD or clozapine followed by AMPH or MK-801 on PPI and global DNA methylation changes in the ventral striatum and prefrontal cortex (PFC); and ii). if the acute antipsychotic-like effects of CBD would last for 24-h. AMPH (5 mg/kg) and MK-801 (0.5 mg/kg) impaired PPI. CBD (30 and 60 mg/kg), similar to clozapine (5 mg/kg), attenuated AMPH- and MK801-induced PPI disruption. AMPH, but not MK-801, increased global DNA methylation in the ventral striatum, an effect prevented by CBD. CBD and clozapine increased, by themselves, DNA methylation in the prefrontal cortex. The acute effects of CBD (30 or 60 mg/kg) on the PPI impairment induced by AMPH or MK-801 was also detectable 24 h later. Altogether, the results show that CBD induces acute antipsychotic-like effects that last for 24-h. It also modulates DNA methylation in the ventral striatum, suggesting a new potential mechanism for its antipsychotic-like effects.


Cannabidiol/pharmacology , Clozapine/pharmacology , Dizocilpine Maleate/pharmacology , Epigenesis, Genetic/drug effects , Prepulse Inhibition/drug effects , Sensory Gating/drug effects , Ventral Striatum/drug effects , Amphetamine/pharmacology , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , DNA Methylation , Dizocilpine Maleate/administration & dosage , Hallucinogens/pharmacology , Male , Mice , Neuroprotective Agents/pharmacology , Prefrontal Cortex/drug effects , Reflex, Startle/drug effects , Schizophrenia , Time Factors
8.
Mol Neurobiol ; 58(2): 777-794, 2021 Feb.
Article En | MEDLINE | ID: mdl-33025509

BACKGROUND: Stress increases DNA methylation, primarily a suppressive epigenetic mechanism catalyzed by DNA methyltransferases (DNMT), and decreases the expression of genes involved in neuronal plasticity and mood regulation. Despite chronic antidepressant treatment decreases stress-induced DNA methylation, it is not known whether inhibition of DNMT would convey rapid antidepressant-like effects. AIM: This work tested such a hypothesis and evaluated whether a behavioral effect induced by DNMT inhibitors (DNMTi) corresponds with changes in DNA methylation and transcript levels in genes consistently associated with the neurobiology of depression and synaptic plasticity (BDNF, TrkB, 5-HT1A, NMDA, and AMPA). METHODS: Male Wistar rats received intraperitoneal (i.p.) injection of two pharmacologically different DNMTi (5-AzaD 0.2 and 0.6 mg/kg or RG108 0.6 mg/kg) or vehicle (1 ml/kg), 1 h or 7 days before the learned helplessness test (LH). DNA methylation in target genes and the correspondent transcript levels were measured in the hippocampus (HPC) and prefrontal cortex (PFC) using meDIP-qPCR. In parallel separate groups, the antidepressant-like effect of 5-AzaD and RG108 was investigated in the forced swimming test (FST). The involvement of cortical BDNF-TrkB-mTOR pathways was assessed by intra-ventral medial PFC (vmPFC) injections of rapamycin (mTOR inhibitor), K252a (TrkB receptor antagonist), or vehicle (0.2 µl/side). RESULTS: We found that both 5-AzaD and RG108 acutely and 7 days before the test decreased escape failures in the LH. LH stress increased DNA methylation and decreased transcript levels of BDNF IV and TrkB in the PFC, effects that were not significantly attenuated by RG108 treatment. The systemic administration of 5-AzaD (0.2 mg/kg) and RG108 (0.2 mg/kg) induced an antidepressant-like effect in FST, which was, however, attenuated by TrkB and mTOR inhibition into the vmPFC. CONCLUSION: These findings suggest that acute inhibition of stress-induced DNA methylation promotes rapid and sustained antidepressant effects associated with increased BDNF-TrkB-mTOR signaling in the PFC.


Antidepressive Agents/pharmacology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/genetics , Gene Expression Regulation , Neuronal Plasticity/genetics , Prefrontal Cortex/physiology , Animals , Behavior, Animal , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Helplessness, Learned , Male , Neuronal Plasticity/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Receptor, trkB/genetics , Receptor, trkB/metabolism , Signal Transduction/drug effects , Stress, Psychological/metabolism , TOR Serine-Threonine Kinases/metabolism
9.
Behav Brain Res ; 388: 112627, 2020 06 18.
Article En | MEDLINE | ID: mdl-32348868

Cannabidiol (CBD), a non-psychotomimetic component of Cannabis sativa plant, shows therapeutic potential in psychiatric disorders, including depression. The molecular mechanisms underlying the antidepressant-like effects of CBD are not yet understood. Previous studies in differentiated skin cells demonstrated that CBD regulates DNA methylation, an overall repressive epigenetic mechanism. Both stress exposure and antidepressant treatment can modulate DNA methylation in the brain, and lead to gene expression changes associated with depression neurobiology. We investigated herein if the antidepressant effect of CBD could be associated with changes in DNA methylation in the prefrontal cortex (PFC) and hippocampus (HPC) of mice submitted to the forced swimming test (FST). Therefore, we assessed: i) the behavioral effects induced by CBD and DNA methylation inhibitors (DNMTi: 5-AzaD and RG108), alone or in association; ii) the effects induced by CBD and DNMTi in global DNA methylation and DNMT activity, in PFC and HPC. Results showed that treatment with CBD (10 mg/kg), 5-AzaD and RG108 (0.2 mg/kg) induced an antidepressant-like effect in the FST. Similar effects were observed after the combination of sub-effective doses of CBD (7 mg/kg) and 5-AzaD or CBD (7 mg/kg) and RG108 (0.1 mg/kg). Also, stress reduced DNA methylation and DNMT activity in the HPC and increased it in the PFC. CBD and DNMTi treatment prevented these changes in both brain structures. Altogether, our results indicate that CBD regulates DNA methylation in brain regions relevant for depression neurobiology, suggesting that this mechanism could be related to CBD-induced antidepressant effects.


Antidepressive Agents/administration & dosage , Cannabidiol/administration & dosage , DNA Methylation , Depression/metabolism , Hippocampus/drug effects , Prefrontal Cortex/drug effects , Animals , Disease Models, Animal , Epigenesis, Genetic/drug effects , Hippocampus/metabolism , Male , Mice , Prefrontal Cortex/metabolism
10.
Behav Pharmacol ; 31(4): 333-342, 2020 06.
Article En | MEDLINE | ID: mdl-31860564

DNA methylation, an epigenetic modification that mediates gene silencing, has been shown to play a role in the neurobiology of major depression. Studies suggested that terpenes inhibit DNA methylation and increase gene expression. The present study investigated the involvement of DNA methylation in the antidepressant-like activity of diene valepotriates, non-glicosilated carbocyclic iridoids that comprise a family of terpenes obtained from Valeriana glechomifolia. The antidepressant-like effect of diene valepotriates acute administration (5 mg/kg, p.o.) in mice submitted to the forced swimming test was followed by a decrease in global DNA methylation in animals' hippocampus (but not in the pre-frontal cortex). Mice pretreatment with anysomicin (a protein synthesis inhibitor) and K252a (an inhibitor of Trk receptors) attenuated diene valepotriates-induced antidepressant-like effect in the forced swimming test. Diene valepotriates elicited an upregulation in the TrkB receptor and a tendency to increase BDNF levels in mice hippocampus. These results demonstrate that DNA methylation could be an in vivo molecular target of diene valepotriates. The diene valepotriates-triggered reduction in hippocampal DNA methylation is accompanied by increased protein synthesis, which is involved in its antidepressant-like activity. Furthermore, BDNF-mediated TrkB signaling may contribute for diene valepotriates antidepressant-like effect.


DNA Methylation/drug effects , Hippocampus/metabolism , Iridoids/pharmacology , Plant Extracts/pharmacology , Receptor, trkB/biosynthesis , Valerian/chemistry , Animals , Anisomycin/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Carbazoles/pharmacology , Indole Alkaloids/pharmacology , Iridoids/antagonists & inhibitors , Male , Mice , Plant Extracts/chemistry , Prefrontal Cortex/metabolism , Up-Regulation/drug effects
11.
Mol Neurobiol ; 56(2): 1070-1081, 2019 Feb.
Article En | MEDLINE | ID: mdl-29869197

Currently available antidepressants have a substantial time lag to induce therapeutic response and a relatively low efficacy. The development of drugs that addresses these limitations is critical to improving public health. Cannabidiol (CBD), a non-psychotomimetic component of Cannabis sativa, is a promising compound since it shows large-spectrum therapeutic potential in preclinical models and humans. However, its antidepressant properties have not been completely investigated. Therefore, the aims of this study were to investigate in male rodents (i) whether CBD could induce rapid and sustained antidepressant-like effects after a single administration and (ii) whether such effects could be related to changes in synaptic proteins/function. Results showed that a single dose of CBD dose-dependently induced antidepressant-like effect (7-30 mg/kg) in Swiss mice submitted to the forced swim test (FST), 30 min (acute) or 7 days (sustained) following treatment. Similar effects were observed in the Flinders Sensitive and Flinders Resistant Line (FSL/FRL) rats and the learned helplessness (LH) paradigm using Wistar rats. The acute antidepressant effects (30 min) were associated with increased expression of synaptophysin and PSD95 in the medial prefrontal cortex (mPFC) and elevated BDNF levels in both mPFC and hippocampus (HPC). CBD also increased spine density in the mPFC after 30 min, but not 7 days later. Intracerebroventricular injection of the TrkB antagonist, K252a (0.05 nmol/µL), or the mTOR inhibitor, rapamycin (1 nmol/µL), abolished the behavioral effects of CBD. These results indicate that CBD induces fast and sustained antidepressant-like effect in distinct animal models relevant for depression. These effects may be related to rapid changes in synaptic plasticity in the mPFC through activation of the BDNF-TrkB signaling pathway. The data support a promising therapeutic profile for CBD as a new fast-acting antidepressant drug.


Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/drug effects , Cannabidiol/pharmacology , Prefrontal Cortex/drug effects , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Hippocampus/metabolism , Male , Mice , Prefrontal Cortex/metabolism , Signal Transduction/drug effects
12.
J Psychopharmacol ; 32(8): 922-931, 2018 08.
Article En | MEDLINE | ID: mdl-29968502

BACKGROUND: Cannabidiol is a non-psychotomimetic compound with antidepressant-like effects. However, the mechanisms and brain regions involved in cannabidiol effects are not yet completely understood. Brain-derived neurotrophic factor/tropomyosin-receptor kinase B/mammalian target of rapamycin (BDNF-TrkB-mTOR) signaling, especially in limbic structures, seems to play a central role in mediating the effects of antidepressant drugs. AIM: Since it is not yet known if BDNF-TrkB-mTOR signaling in the hippocampus is critical to the antidepressant-like effects of cannabidiol, we investigated the effects produced by cannabidiol (10/30/60 nmol/0.2 µL) micro-injection into the hippocampus of mice submitted to the forced swim test and to the open field test. METHODS: Independent groups received intra-hippocampal injections of rapamycin (mTOR inhibitor, 0.2 nmol/0.2 µL) or K252 (Trk antagonist, 0.01 nmol/0.2 µL), before the systemic (10 mg/kg) or hippocampal (10 nmol/0.2µL) injection of cannabidiol, and were submitted to the same tests. BDNF levels were analyzed in the hippocampus of animals treated with cannabidiol (10 mg/kg). RESULTS: Systemic cannabidiol administration induced antidepressant-like effects and increased BDNF levels in the dorsal hippocampus. Rapamycin, but not K252a, injection into the dorsal hippocampus prevented the antidepressant-like effect induced by systemic cannabidiol treatment (10 mg/kg). Differently, hippocampal administration of cannabidiol (10 nmol/0.2 µL) reduced immobility time, an effect that was blocked by both rapamycin and K252a local microinjection. CONCLUSION: Altogether, our data suggest that the hippocampal BDNF-TrkB-mTOR pathway is vital for cannabidiol-induced antidepressant-like effect when the drug is locally administered. However, other brain regions may also be involved in cannabidiol-induced antidepressant effect upon systemic administration.


Behavior, Animal/drug effects , Cannabidiol/adverse effects , Hippocampus/drug effects , Hippocampus/metabolism , Stress, Psychological/chemically induced , Stress, Psychological/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Carbazoles/pharmacology , Depression/chemically induced , Depression/drug therapy , Depression/metabolism , Indole Alkaloids/pharmacology , Male , Mice , Receptor, trkB/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Swimming
13.
Article En | MEDLINE | ID: mdl-29885468

Cannabidiol (CBD) is a compound of Cannabis sativa with relevant therapeutic potential in several neuropsychiatric disorders including depression. CBD treatment has shown significant antidepressant-like effects in different rodent preclinical models. However, the mechanisms involved in CBD-induced antidepressant effects are still poorly understood. Therefore, this work aimed at investigating the participation of serotonin (5-HT) and/or noradrenaline (NA) in CBD-induced antidepressant-like effects in the forced swimming test (FST) by: 1) testing if CBD co-administration with serotonergic (fluoxetine, FLX) or noradrenergic (desipramine, DES) antidepressants would have synergistic effects; and 2) investigating if 5-HT or NA depletion would impair CBD-induced behavioral effects. Results showed that CBD (10 mg/kg), FLX (10 mg/kg) and DES (5 mg/kg) induced antidepressant-like effects in mice submitted to FST. Ineffective doses of CBD (7 mg/kg), when co-administered with ineffective doses of FLX (5 mg/kg) or DES (2.5 mg/kg) resulted in significant antidepressant-like effects, thus implicating synergistic and/or additive mechanisms. Pretreatment with PCPA (an inhibitor of serotonin synthesis: 150 mg/kg, i.p., once per day for 4 days), but not DSP-4 (a noradrenergic neurotoxin: 1 µg/µl, i.c.v., 24 h before the test), reduced monoamine levels in the brain. However, only PCPA treatment abolished CBD-induced behavioral effects in FST, indicating the participation of serotonergic mechanisms. None of the treatments induced locomotor effects. Our results suggest that the antidepressant-like effect induced by CBD in the FST is dependent on serotonin levels in the central nervous system (CNS).


Antidepressive Agents/pharmacology , Brain/drug effects , Cannabidiol/pharmacology , Depressive Disorder/drug therapy , Serotonin/metabolism , Animals , Brain/metabolism , Depressive Disorder/metabolism , Desipramine/pharmacology , Disease Models, Animal , Drug Synergism , Fluoxetine/pharmacology , Male , Mice , Norepinephrine/metabolism , Random Allocation
14.
Behav Brain Res ; 343: 8-15, 2018 05 02.
Article En | MEDLINE | ID: mdl-29378290

Stress and antidepressant treatment can modulate DNA methylation in promoter region of genes related to neuroplasticity and mood regulation, thus implicating this epigenetic mechanism in depression neurobiology and treatment. Accordingly, systemic administration of DNA methyltransferase (DNMT) inhibitors induces antidepressant-like effects in rodents. DNA methylation is conveyed by DNMT 1, 3a and 3b isoforms, which are differentially expressed in the brain. In order to investigate if the behavioral effects of antidepressants could be associated with changes in DNA methylation and DNMT expression, we investigated the effects induced by acute and repeated antidepressant treatment on DNA methylation and DNMT expression (1, 3a and 3b isoforms) in different brain regions of rats exposed to a stress model of depression, the learned helplessness (LH). Therefore, rats were exposed to pretest and treated with one or seven injections of vehicle or imipramine (15 mg kg-1), with test session performed one hour after the last injection. Chronic, but not acute, imipramine administration attenuated escape failures during the test, a well described antidepressant-like effect in this model. DNA methylation and DNMT (1, 3a and 3b) levels were measured in the dorsal and ventral hippocampus (dHPC, vHPC) and in the prefrontal cortex (PFC) of rats exposed to stress and treatment. Stress increased DNA methylation, DNMT3a and DNMT3b expression in the dHPC and PFC. Chronic, but not acute, imipramine administration attenuated stress effects only in the PFC. These results suggest the regulation of DNA methylation in the PFC may be an important mechanism for antidepressant-like effects in the LH model.


Antidepressive Agents/pharmacology , DNA Methylation/drug effects , DNA Modification Methylases/metabolism , Depressive Disorder/drug therapy , Hippocampus/drug effects , Prefrontal Cortex/drug effects , Animals , DNA Methylation/physiology , Depressive Disorder/genetics , Depressive Disorder/metabolism , Disease Models, Animal , Electroshock , Gene Expression/drug effects , Helplessness, Learned , Hippocampus/metabolism , Imipramine/pharmacology , Isoenzymes/metabolism , Male , Prefrontal Cortex/metabolism , Random Allocation , Rats, Wistar , Stress, Psychological/drug therapy , Stress, Psychological/genetics , Stress, Psychological/metabolism
15.
Front Pharmacol ; 8: 269, 2017.
Article En | MEDLINE | ID: mdl-28588483

Beneficial effects of cannabidiol (CBD) have been described for a wide range of psychiatric disorders, including anxiety, psychosis, and depression. The mechanisms responsible for these effects, however, are still poorly understood. Similar to clinical antidepressant or atypical antipsychotic drugs, recent findings clearly indicate that CBD, either acutely or repeatedly administered, induces plastic changes. For example, CBD attenuates the decrease in hippocampal neurogenesis and dendrite spines density induced by chronic stress and prevents microglia activation and the decrease in the number of parvalbumin-positive GABA neurons in a pharmacological model of schizophrenia. More recently, it was found that CBD modulates cell fate regulatory pathways such as autophagy and others critical pathways for neuronal survival in neurodegenerative experimental models, suggesting the potential benefit of CBD treatment for psychiatric/cognitive symptoms associated with neurodegeneration. These changes and their possible association with CBD beneficial effects in psychiatric disorders are reviewed here.

16.
Curr Top Med Chem ; 17(7): 771-793, 2017.
Article En | MEDLINE | ID: mdl-27086781

Neuronal network and plasticity change as a function of experience. Altered neural connectivity leads to distinct transcriptional programs of neuronal plasticity-related genes. The environmental challenges throughout life may promote long-lasting reprogramming of gene expression and the development of brain disorders. The modifications in neuronal epigenome mediate gene-environmental interactions and are required for activity-dependent regulation of neuronal differentiation, maturation and plasticity. Here, we highlight the latest advances in understanding the role of the main players of epigenetic machinery (DNA methylation and demethylation, histone modifications, chromatin-remodeling enzymes, transposons, and non-coding RNAs) in activity-dependent and long- term neural and synaptic plasticity. The review focuses on both the transcriptional and post-transcriptional regulation of gene expression levels, including the processes of promoter activation, alternative splicing, regulation of stability of gene transcripts by natural antisense RNAs, and alternative polyadenylation. Further, we discuss the epigenetic aspects of impaired neuronal plasticity and the pathogenesis of neurodevelopmental (Rett syndrome, Fragile X Syndrome, genomic imprinting disorders, schizophrenia, and others), stressrelated (mood disorders) and neurodegenerative Alzheimer's, Parkinson's and Huntington's disorders. The review also highlights the pharmacological compounds that modulate epigenetic programming of gene expression, the potential treatment strategies of discussed brain disorders, and the questions that should be addressed during the development of effective and safe approaches for the treatment of brain disorders.


Epigenesis, Genetic , Neuronal Plasticity , Humans , RNA Processing, Post-Transcriptional , RNA, Untranslated/genetics , Transcription, Genetic
17.
Behav Pharmacol ; 28(1): 19-29, 2017 02.
Article En | MEDLINE | ID: mdl-27779493

Systemic or hippocampal administration of nitric oxide (NO) synthase inhibitors induces antidepressant-like effects in animals, implicating increased hippocampal levels of NO in the neurobiology of depression. However, the role played by different NO synthase in this process has not been clearly defined. As stress is able to induce neuroinflammatory mechanisms and trigger the expression of inducible nitric oxide synthase (iNOS) in the brain, as well as upregulate neuronal nitric oxide synthase (nNOS) activity, the aim of the present study was to investigate the possible differential contribution of hippocampal iNOS and nNOS in the modulation of the consequences of stress elicited by the forced swimming test. Male Wistar rats received intrahippocampal injections, immediately after the pretest or 1 h before the forced swimming test, of selective inhibitors of nNOS (N-propyl-L-arginine), iNOS (1400W), or sGC (ODQ), the main pharmacological target for NO. Stress exposure increased nNOS and phospho-nNOS levels at all time points, whereas iNOS expression was increased only 24 h after the pretest. All drugs induced an antidepressant-like effect. However, whereas the nNOS inhibitor was equally effective when injected at different times, the iNOS inhibitor was more effective 24 h after the pretest. These results suggest that hippocampal nNOS and iNOS contribute to increase in NO levels in response to stress, although with a differential time course after stress exposure.


Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Soluble Guanylyl Cyclase/metabolism , Stress, Psychological/metabolism , Amidines/pharmacology , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Benzylamines/pharmacology , Disease Models, Animal , Hippocampus/metabolism , Male , Nitric Oxide/metabolism , Oxadiazoles/pharmacology , Rats , Rats, Wistar , Swimming , Time Factors
18.
Behav Pharmacol ; 25(3): 187-96, 2014 Jun.
Article En | MEDLINE | ID: mdl-24710314

Systemic as well as hippocampal administration of nNOS inhibitors induces antidepressant-like effects in animal models. However, the mechanisms underlying these effects have not been completely understood. Evidence has suggested that nNOS inhibition increases serotonin signaling in the brain. Moreover, activation of prosencephalic 5HT1A receptors is considered to mediate stress coping and antidepressant effects. On this basis, the aim of this study was to investigate the hypothesis that the antidepressant-like effect induced by nNOS inhibition in the dorsal hippocampus (DH) would involve local serotonergic signaling and 5HT1A receptor activation. Therefore, rats were subjected to the forced swimming test and received microinjections of fluoxetine, NPA (Nω-propyl-L-arginine, selective nNOS inhibitor), WAY100635 (5HT1A antagonist), and vehicle, alone or in combination, into the DH. Exposure to the forced swimming test increased nitric oxide acid levels in the DH. The administration of NPA or fluoxetine in the DH induced dose-dependent antidepressant-like effects. WAY100635 microinjection in the DH did not induce any effect per se, but it counteracted NPA-induced and fluoxetine-induced effects. These results suggest that the antidepressant-like effect induced by the administration of nNOS inhibitors in the DH involves local serotonergic signaling and 5HT1A receptor activation.


Antidepressive Agents/pharmacology , Hippocampus/drug effects , Hippocampus/enzymology , Nitric Oxide Synthase Type I/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Exploratory Behavior/drug effects , Fluoxetine/pharmacology , Male , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Serotonin Antagonists/pharmacology , Stress, Psychological/drug therapy , Swimming/psychology
19.
Br J Pharmacol ; 164(6): 1711-21, 2011 Nov.
Article En | MEDLINE | ID: mdl-21585346

BACKGROUND AND PURPOSE: Epigenetic modifications are thought to play an important role in the neurobiology of depression. Antidepressant treatment induces histone acetylation in the hippocampus, which is associated with transcriptional activation, whereas stress increases DNA methylation, which is associated with transcriptional repression. Because the specific involvement of DNA methylation in the regulation of depressive-like behaviours is not yet known, we have investigated the effects induced by systemic or intra-hippocampal administration of inhibitors of DNA methyltransferase (DNMT) in rats submitted to a range of behavioural tests. EXPERIMENTAL APPROACH: Rats received i.p. injections of 5-aza-2-deoxycytidine (5-azaD, 0.1-0.8 mg·kg(-1) ), 5-azacytidine (5-azaC, 0.4-3.2 mg·kg(-1) ), imipramine (15 mg·kg(-1) ) or vehicle and were submitted to the forced swimming test (FST) or open field test (OFT). Other groups of rats received intra-hippocampal injection of DNMT inhibitors. KEY RESULTS: Systemic administration of DNMT inhibitors induced a dose-dependent antidepressant-like effect, which was followed by decreased DNA methylation and increased brain-derived neurotrophic factor (BDNF) levels in the hippocampus. Hippocampal inhibition of DNA methylation induced similar behavioural effects. No treatment induced any locomotor effects in the OFT. Antidepressant-like effects of 5-azaD were confirmed in mice submitted to the FST or the tail suspension test. CONCLUSIONS AND IMPLICATIONS: Systemic, as well as hippocampal, inhibition of DNA methylation induced antidepressant-like effects. These effects could be associated with increased hippocampal expression of BDNF. Our data give further support to the hypothesis that DNA methylation is an important epigenetic mechanism involved in the development of depressive-like behaviours.


Antidepressive Agents/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Behavior, Animal/drug effects , DNA Methylation/drug effects , Depression/drug therapy , Hippocampus/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Decitabine , Depression/metabolism , Disease Models, Animal , Hindlimb Suspension , Hippocampus/metabolism , Imipramine/pharmacology , Male , Mice , Rats , Swimming
...