Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Pharmacol Biochem Behav ; 220: 173468, 2022 10.
Article in English | MEDLINE | ID: mdl-36174752

ABSTRACT

Major depressive disorder (MDD) is the most prevalent mood disorder globally. Most antidepressants available for the treatment of MDD increase the concentration of monoamines in the synaptic cleft. However, such drugs have a high latency time to obtain benefits. Thus, new antidepressants with fast action and robust efficacy are very important. This study evaluated the effects of escitalopram, ketamine, and probiotic Bifidobacterium infantis in rats submitted to the maternal deprivation (MD). MD rats received saline, escitalopram, ketamine, or probiotic for 10, 30, or 50 days, depending on the postnatal day (PND):21, 41, and 61. Following behavior, this study examined the integrity of the blood-brain barrier (BBB) and oxidative stress markers. MD induced depressive-like behavior in females with PND21 and males with PND61. All treatments reversed depressive-like behavior in females and escitalopram and ketamine in males. MD induced an increase in the permeability of the BBB, an imbalance between oxidative stress and antioxidant defenses. Treatments regulated the oxidative damage and the integrity of the BBB induced by MD. The treatment with escitalopram, ketamine, or probiotics may prevent behavioral and neurochemical changes associated with MDD, depending on the developmental period and gender.


Subject(s)
Antidepressive Agents , Depressive Disorder, Major , Sex Characteristics , Stress, Psychological , Animals , Female , Male , Rats , Antidepressive Agents/therapeutic use , Antioxidants/metabolism , Depressive Disorder, Major/drug therapy , Ketamine , Rats, Wistar , Stress, Psychological/drug therapy , Escitalopram
2.
Brain Res Bull ; 172: 129-138, 2021 07.
Article in English | MEDLINE | ID: mdl-33932489

ABSTRACT

This study aimed to evaluate the effects of ketamine, on behavioral parameters, oxidative stress, and inflammation in the brain of male and female rats submitted to the animal model of maternal deprivation (MD). Wistar rats were deprived of maternal care in the first 10 days of life (three hours daily). As adults, male and female rats were divided: control + saline deprived + saline and deprived + ketamine (15 mg/kg). The behavior was evaluated through the open field and forced swimming tests. Then brain was removed for analysis of oxidative damage, the activity of superoxide dismutase (SOD), catalase (CAT), and myeloperoxidase (MPO) activity, and levels of interleukin-6 (IL-6). MD induced depressive behavior in males and ketamine reversed these changes. MD induced an increase in lipid peroxidation in males and females; ketamine reversed these effects in males. Protein carbonylation was increased in males and females, with ketamine decreasing such effects. The concentration of nitrite/nitrate increased in males and females, whereas ketamine decreased this in the PFC of males. SOD and CAT activities were decreased in male and female deprived groups and deprived groups treated with ketamine. MPO activity and IL-6 levels increased in males subjected to MD and ketamine reversed this effect. The results suggest that stressful events in early life can induce behavioral, neuroimmune changes, and oxidative stress, however, such effects depend on sex and brain area. Ketamine presents anti-inflammatory and antioxidant properties and could be considered an alternative for individuals who are resistant to classical treatments.


Subject(s)
Behavior, Animal/drug effects , Hippocampus/drug effects , Ketamine/pharmacology , Maternal Deprivation , Oxidative Stress/drug effects , Prefrontal Cortex/drug effects , Stress, Psychological/metabolism , Animals , Catalase/metabolism , Female , Hippocampus/metabolism , Interleukin-6/metabolism , Male , Malondialdehyde/metabolism , Motor Activity/drug effects , Peroxidase/metabolism , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Sex Factors , Superoxide Dismutase/metabolism
3.
Brain Res Bull ; 171: 196-208, 2021 06.
Article in English | MEDLINE | ID: mdl-33838211

ABSTRACT

Sex differences are often observed in psychiatric patients, especially major depressive disorders (MDD), schizophrenia, and developmental disorders, including autism spectrum disorders (ASDs). The prevalence rates between males and females seem variate according to the clinical condition. Although the findings are still incipient, it is suggested that these differences can involve neuroanatomical, neurochemical, and physiological sex differences. In this context, the microbiota-gut-brain axis hypothesis arises to explain some aspects of the complex pathophysiology of neuropsychiatric disorders. The microbiota composition is host-specific and can change conforming to age, sex, diet, medication, exercise, and others. The communication between the brain and the gut is bidirectional and may impact the entire system homeostasis. Many pathways appear to be involved, including neuroanatomic communication, neuroendocrine pathways, immune system, bacteria-derived metabolites, hormones, neurotransmitters, and neurotrophic factors. Although the clinical and preclinical studies are sparse and not very consistent, they suggest that sex differences in the gut microbiota may play an essential role in some neuropsychiatric conditions. Thus, this narrative review has as a mainly aim to show the points sex-related patterns associated to the gut-microbiota-brain axis in the MDD, ASDs, and schizophrenia.


Subject(s)
Brain-Gut Axis/physiology , Gastrointestinal Microbiome/physiology , Mental Disorders/metabolism , Animals , Female , Humans , Male , Mental Disorders/microbiology
4.
Eur J Neurosci ; 53(1): 222-235, 2021 01.
Article in English | MEDLINE | ID: mdl-31785168

ABSTRACT

Major depressive disorder (MDD) is a psychiatric condition that affects a large number of people in the world, and the treatment existents do not work for all individuals affected. Thus, it is believed that other systems or pathways which regulate brain networks involved in mood regulation and cognition are associated with MDD pathogenesis. Studies in humans and animal models have been shown that in MDD there are increased levels of inflammatory mediators, including cytokines and chemokines in both periphery and central nervous system (CNS). In addition, microglial activation appears to be a key event that triggers changes in signaling cascades and gene expression that would be determinant for the onset of depressive symptoms. Recent researches also point out that changes in the gut microbiota would lead to a systemic inflammation that in different ways would reach the CNS modulating inflammatory pathways and especially the microglia, which could influence responses to treatments. Moreover, pre- and probiotics have shown antidepressant responses and anti-inflammatory effects. This review will focus on studies that show the relationship of inflammation with the gut microbiota-brain axis and its relation with MDD.


Subject(s)
Depressive Disorder, Major , Gastrointestinal Microbiome , Probiotics , Animals , Brain , Depression , Depressive Disorder, Major/therapy , Humans
5.
Oxid Med Cell Longev ; 2019: 8637970, 2019.
Article in English | MEDLINE | ID: mdl-30944699

ABSTRACT

Both conditions, major depressive disorder (MDD) and diabetes mellitus (DM) are chronic and disabling diseases that affect a very significant percentage of the world's population. Studies have been shown that patients with DM are more susceptible to develop depression, when compared to the general population. The opposite also happens; MDD could be a risk factor for DM development. Some mechanisms have been proposed to explain the pathophysiological mechanisms involved with these conditions, such as excess of glucocorticoids, hyperglycemia, insulin resistance, and inflammation. These processes can lead to an increase in damage to biomolecules and a decrease in antioxidant defense capacity, leading to oxidative stress.


Subject(s)
Diabetes Mellitus/psychology , Oxidative Stress/physiology , Depressive Disorder, Major , Diabetes Complications , Diabetes Mellitus/pathology , Female , Humans , Male
6.
Pharmacol Rep ; 68(1): 177-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26721370

ABSTRACT

BACKGROUND: Ketamine, an antagonist of N-methyl-d-aspartate (NMDA) receptors, has presented antidepressant effects in basic and clinical studies. The MAPK kinase (MEK) signaling pathway could be a target for novel antidepressant drugs and an important pathway involved in neuronal plasticity. Thus, this study evaluated the effects of the administration of ketamine on the phosphorylation of TrKB and CREB, and oxidative stress parameters in the prefrontal cortex (PFC), hippocampus, amygdala, and nucleus accumbens (NAc) rats, after the inhibition of MAPK pathway (PD184161). METHODS: Male adult Wistar rats were submitted to a surgical procedure to receive a single dose of a pharmacological inhibitor of MAPK (PD184161) at a dose of (0.1µg/µl) or vehicle. Then, they were divided: 1) vehicle+saline; 2) inhibitor PD184161+saline; 3) vehicle+ketamine 15mg/kg; and 4) inhibitor PD184161+ketamine 15mg/kg. RESULTS: MEK inhibitor and ketamine increased the phosphorylation of the transcription factor cAMP response element-binding protein (pCREB) and neurotrophic factor/tropomyosin related kinase B receptor (pTrKB) in the PFC, and decreased pCREB in the hippocampus. The MEK inhibitor abolished ketamine's effects in the hippocampus. In the amygdala, pCREB was decreased, and pTrKB was increased after MEK inhibitor plus ketamine. Ketamine increased the thiobarbituric acid reactive species (TBARS) in the PFC, hippocampus, amygdala, and NAc; MEK inhibitor antagonized these effects. The carbonyl was increased in the PFC by both ketamine and MEK inhibitor, but inhibitor infusion plus ketamine administration reduced this effect. In the amygdala, MEK inhibitor increased carbonyl. CONCLUSION: Ketamine's effects on pCREB, pTrKB, and oxidative stress are mediated, at least in part, by a mechanism dependent of MAPK signaling inhibition.


Subject(s)
Aniline Compounds/administration & dosage , Benzamides/administration & dosage , Cyclic AMP Response Element-Binding Protein/metabolism , Ketamine/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Oxidative Stress/physiology , Receptor, trkB/metabolism , Animals , Brain/drug effects , Brain/metabolism , Infusions, Intravenous , Male , Oxidative Stress/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Wistar
7.
Diabetes Metab Res Rev ; 32(3): 278-88, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26432993

ABSTRACT

Studies have shown a relationship between diabetes mellitus (DM) and the development of major depressive disorder. Alterations in oxidative stress are associated with the pathophysiology of both diabetes mellitus and major depressive disorder. This study aimed to evaluate the effects of antioxidants N-acetylcysteine and deferoxamine on behaviour and oxidative stress parameters in diabetic rats. To this aim, after induction of diabetes by a single dose of alloxan, Wistar rats were treated with N-acetylcysteine or deferoxamine for 14 days, and then depressive-like behaviour was evaluated. Oxidative stress parameters were assessed in the prefrontal cortex, hippocampus, amygdala, nucleus accumbens and pancreas. Diabetic rats displayed depressive-like behaviour, and treatment with N-acetylcysteine reversed this alteration. Carbonyl protein levels were increased in the prefrontal cortex, hippocampus and pancreas of diabetic rats, and both N-acetylcysteine and deferoxamine reversed these alterations. Lipid damage was increased in the prefrontal cortex, hippocampus, amygdala and pancreas; however, treatment with N-acetylcysteine or deferoxamine reversed lipid damage only in the hippocampus and pancreas. Superoxide dismutase activity was decreased in the amygdala, nucleus accumbens and pancreas of diabetic rats. In diabetic rats, there was a decrease in catalase enzyme activity in the prefrontal cortex, amygdala, nucleus accumbens and pancreas, but an increase in the hippocampus. Treatment with antioxidants did not have an effect on the activity of antioxidant enzymes. In conclusion, animal model of diabetes produced depressive-like behaviour and oxidative stress in the brain and periphery. Treatment with antioxidants could be a viable alternative to treat behavioural and biochemical alterations induced by diabetes.


Subject(s)
Antioxidants/pharmacology , Brain/drug effects , Depressive Disorder/prevention & control , Diabetes Mellitus, Experimental/drug therapy , Oxidative Stress/drug effects , Pancreas/drug effects , Acetylcysteine/pharmacology , Animals , Behavior, Animal/drug effects , Brain/metabolism , Brain/pathology , Deferoxamine/pharmacology , Depressive Disorder/metabolism , Depressive Disorder/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/psychology , Free Radical Scavengers/pharmacology , Lipid Peroxidation/drug effects , Male , Pancreas/metabolism , Pancreas/pathology , Rats , Rats, Wistar , Siderophores/pharmacology
8.
Curr Neurovasc Res ; 12(3): 283-92, 2015.
Article in English | MEDLINE | ID: mdl-26036973

ABSTRACT

Several studies have found that the molecular mechanisms of mitochondrial energy metabolism are impaired in major depressive disorder (MDD). Classic antidepressants and atypical antipsychotics can alter the function of enzymes involved in adenosine triphosphate (ATP) metabolism. Quetiapine is an atypical antipsychotic that, in addition to having a therapeutic benefit in treating MDD, appears to exert antioxidant and neuroprotective effects. Therefore, we aimed to evaluate the acute and chronic effects of quetiapine on the activity of enzyme complexes I to IV of the mitochondrial respiratory chain and creatine kinase (CK) in brain regions involved with MDD. After a single dose or serial injections over 14 days of quetiapine (20, 40, and 80 mg) were administered, isolates from the pre- frontal cortex, hippocampus, amygdala and nucleus accumbens were analyzed for enzyme activity levels. The enzyme activity varied according to the dose, brain region, and acute or chronic dosing protocols. In general, complexes I-III activity was increased, especially after acute administration. Acute administration also increased the activity of complex IV and CK in the amygdala while complex I was inhibited in the prefrontal cortex and nucleus accumbens. These results suggest that quetiapine produces an increase in respiratory chain complex activity, which may be underlying its efficacy against psychiatric disorders and neuronal damage.


Subject(s)
Antipsychotic Agents/pharmacology , Brain , Multienzyme Complexes/metabolism , Quetiapine Fumarate/pharmacology , Analysis of Variance , Animals , Antidepressive Agents, Tricyclic/pharmacology , Brain/anatomy & histology , Brain/drug effects , Brain/enzymology , Creatine Kinase/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Enzymologic/drug effects , Imipramine/pharmacology , Male , Multienzyme Complexes/classification , Rats , Rats, Wistar , Time Factors
9.
Mutat Res ; 772: 30-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25772108

ABSTRACT

This study aimed at investigating the effects of chronic mild stress on DNA damage, NMDA receptor subunits and glutamate transport levels in the brains of rats with an anxious phenotype, which were selected to represent both the high-freezing (CHF) and low-freezing (CLF) lines. The anxious phenotype induced DNA damage in the hippocampus, amygdala and nucleus accumbens (NAc). CHF rats subjected to chronic stress presented a more pronounced DNA damage in the hippocampus and NAc. NMDAR1 were increased in the prefrontal cortex (PC), hippocampus and amygdala of CHF, and decreased in the hippocampus, amygdala and NAc of CHF stressed. NMDAR2A were decreased in the amygdala of the CHF and stressed; and increased in CHF stressed. NMDRA2A in the NAc was increased after stress, and decreased in the CLF. NMDAR2B were increased in the hippocampus of CLF and CHF. In the amygdala, there was a decrease in the NMDAR2B for stress in the CLF and CHF. NMDAR2B in the NAc were decreased for stress and increased in the CHF; in the PC NMDAR2B increased in the CHF. EAAT1 increased in the PC of CLF+stress. In the hippocampus, EAAT1 decreased in all groups. In the amygdala, EAAT1 decreased in the CLF+stress and CHF. EAAT2 were decreased in the PC for stress, and increased in CHF+control. In the hippocampus, the EAAT2 were increased for the CLF and decreased in the CLF+stress. In the amygdala, there was a decrease in the EATT2 in the CLF+stress and CHF. These findings suggest that an anxious phenotype plus stress may induce a more pronounced DNA damage, and promote more alterations in the glutamatergic system. These findings may help to explain, at least in part, the common point of the mechanisms involved with the pathophysiology of depression and anxiety.


Subject(s)
Anxiety/metabolism , Brain/metabolism , DNA Damage , Glutamic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Psychological/metabolism , Animals , Anxiety/genetics , Anxiety/pathology , Brain/pathology , Depression/genetics , Depression/metabolism , Depression/pathology , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/genetics , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/genetics , Species Specificity , Stress, Psychological/genetics , Stress, Psychological/pathology
10.
Dev Neurobiol ; 75(11): 1268-81, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25728399

ABSTRACT

Ketamine, an antagonist of N-methyl-d-aspartate receptors, has produced rapid antidepressant effects in patients with depression, as well as in animal models. However, the extent and duration of the antidepressant effect over longer periods of time has not been considered. This study evaluated the effects of single dose of ketamine on behavior and oxidative stress, which is related to depression, in the brains of adult rats subjected to maternal deprivation. Deprived and nondeprived Wistar rats were divided into four groups nondeprived+saline; nondeprived+S-ketamine (15 mg/kg); deprived+saline; deprived+S-ketamine (15 mg/kg). A single dose of ketamine or saline was administrated during the adult phase, and 14 days later depressive-like behavior was assessed. In addition, lipid damage, protein damage, and antioxidant enzyme activities were evaluated in the rat brain. Maternal deprivation induces a depressive-like behavior, as verified by an increase in immobility and anhedonic behavior. However, a single dose of ketamine was able to reverse these alterations, showing long-term antidepressant effects. The brains of maternally deprived rats had an increase in protein oxidative damage and lipid peroxidation, but administration of a single dose of ketamine reversed this damage. The activities of antioxidant enzymes superoxide dismutase and catalase were reduced in the deprived rat brains. However, ketamine was also able to reverse these changes. In conclusion, these findings indicate that a single dose of ketamine is able to induce long-term antidepressant effects and protect against neural damage caused by oxidative stress in adulthood rats following maternal deprivation.


Subject(s)
Antidepressive Agents/administration & dosage , Brain/drug effects , Depressive Disorder/drug therapy , Ketamine/administration & dosage , Oxidative Stress/drug effects , Anhedonia/drug effects , Anhedonia/physiology , Animals , Brain/physiopathology , Catalase/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Maternal Deprivation , Oxidative Stress/physiology , Rats, Wistar , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology , Superoxide Dismutase/metabolism
11.
Curr Neurovasc Res ; 12(1): 73-84, 2015.
Article in English | MEDLINE | ID: mdl-25613382

ABSTRACT

Studies have suggested that ketamine, a nonselective NMDA receptor antagonist, could be a new drug in the treatment of major depression, but the way ketamine presents such effects remains to be elucidated. Therefore, the objective of this paper was to evaluate the effects of ketamine treatment on parameters related to depression in the brain of adult rats subjected to an animal model of depression. The animals were divided into: non-deprived + saline; non-deprived + ketamine; deprived + saline; deprived + ketamine. Treatments involving ketamine (15 mg/kg) were administered once a day during 14 days in the animal's adult phase. After treatment, the brain derived-neurotrophic factor (BDNF) levels, oxidative stress and energy metabolism activity were evaluated in brain structures of rats involved in the circuit of depression. In the amygdala, hippocampus and nucleus accumbens (NAc), a reduction in BDNF levels was observed in deprived rats, but the animals treated with ketamine reversed the effects of this animal model only in the amygdala and NAc. In addition to this, the complex I activity, in deprived rats, was diminished in the prefrontal cortex (PFC) and amygdala; in the PFC and hippocampus, the complex II-III was diminished in deprived rats; still the administration of ketamine increased the complex IV activity in the PFC and amygdala of rats submitted to the maternal deprivation. In deprived rats, the creatine kinase activity was reduced in the PFC and amygdala, however the administration of ketamine reversed this decrease in the amygdala. The malondialdehyde (MDA) equivalents were increased in non-deprived rats treated with ketamine in the PFC and NAc. Carbonyl levels in the PFC were diminished in control rats that received saline. Though ketamine treatment reversed this effect in deprived rats in the PFC and hippocampus. Still, in NAc, the carbonyl levels were diminished in deprived rats. The superoxide dismutase (SOD) activity was increased in control rats that received ketamine in the PFC and NAc, and were diminished in deprived rats that received saline or ketamine in the PFC and hippocampus. These findings may help to explain that dysfunctions involving BDNF, oxidative stress and energy metabolism within specific brain areas, may be linked with the pathophysiology of depression, and antidepressant effects of ketamine can be positive, at least partially due to the control of these pathways.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Energy Metabolism/drug effects , Excitatory Amino Acid Antagonists/therapeutic use , Ketamine/therapeutic use , Oxidative Stress/drug effects , Analysis of Variance , Animals , Animals, Newborn , Creatine Kinase/metabolism , Depression/pathology , Disease Models, Animal , Female , Male , Maternal Deprivation , Pregnancy , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances
12.
Metab Brain Dis ; 30(2): 545-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25112549

ABSTRACT

Studies have been suggested that minocycline can be a potential new agent for the treatment of depression. In addition, both oxidative stress and energy metabolism present an important role in pathophysiology of depression. So, the present study was aimed to evaluate the effects of minocycline on stress oxidative parameters and energy metabolism in the brain of adult rats submitted to the chronic mild stress protocol (CMS). After CMS Wistar, both stressed animals as controls received twice ICV injection of minocycline (160 µg) or vehicle. The oxidative stress and energy metabolism parameters were assessed in the prefrontal cortex (PF), hippocampus (HIP), amygdala (AMY) and nucleus accumbens (Nac). Our findings showed that stress induced an increase on protein carbonyl in the PF, AMY and NAc, and mynocicline injection reversed this alteration. The TBARS was increased by stress in the PF, HIP and NAc, however, minocycline reversed the alteration in the PF and HIP. The Complex I was incrased in AMY by stress, and minocycline reversed this effect, however reduced Complex I activity in the NAc; Complex II reduced in PF and AMY by stress or minocycline; the Complex II-III increased in the HIP in stress plus minocycline treatment and in the NAc with minocycline; in the PF and HIP there were a reduced in Complex IV with stress and minocycline. The creatine kinase was reduced in AMY and NAc with stress and minocycline. In conclusion, minocycline presented neuroprotector effects by reducing oxidative damage and regulating energy metabolism in specific brain areas.


Subject(s)
Antioxidants/pharmacology , Brain Chemistry/drug effects , Energy Metabolism/drug effects , Minocycline/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Stress, Psychological/metabolism , Animals , Chronic Disease , Creatine Kinase/metabolism , Electron Transport Complex I/drug effects , Electron Transport Complex II/drug effects , Injections, Intraventricular , Male , Rats , Rats, Wistar , Stress, Psychological/drug therapy , Thiobarbituric Acid Reactive Substances/metabolism
13.
Neurosci Lett ; 584: 83-7, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25459283

ABSTRACT

A growing body of evidence points toward an association between the glutamatergic system, as well as immune system dysregulation and major depression. So, the present study was aimed at evaluating the behavioral and molecular effects of the ketamine, an antagonist of the N-methyl-D-aspartate (NMDA) receptor of glutamate in maternally deprived adult rats. In deprived rats treated with saline, we observed an increase in the immobility time; however, ketamine treatment reversed this effect, decreasing immobility time. In addition, maternal deprivation induced an increase in cytokines: TNF-α and IL-1 in serum, and in IL-6 in serum and cerebrospinal fluid (CSF). Interestingly, ketamine treatment reduced the levels of all the cytokines in deprived rats. In conclusion, these findings further support a relationship between immune activation and depression. Considering the action of ketamine, this study suggested that antagonists of the NMDA receptor, such as ketamine, could exert their effects by modulation of the immune system.


Subject(s)
Antidepressive Agents/therapeutic use , Cytokines/blood , Depression/drug therapy , Ketamine/therapeutic use , Maternal Deprivation , Animals , Cytokines/cerebrospinal fluid , Depression/immunology , Depression/psychology , Female , Interleukin-1/blood , Interleukin-6/blood , Interleukin-6/cerebrospinal fluid , Male , Rats, Wistar , Tumor Necrosis Factor-alpha/blood
14.
J Psychiatr Res ; 55: 15-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24819632

ABSTRACT

Studies have pointed to a relationship between MAPK kinase (MEK) signaling and the behavioral effects of antidepressant drugs. So, in the present study we examined the behavioral and molecular effects of ketamine, an antagonist of the N-methyl-d-aspartate receptor (NMDA), which has been shown to have an antidepressant effect after the inhibition of MEK signaling in Wistar rats. Our results showed that acute administration of the MEK inhibitor PD184161, produced depressive-like behavior and stopped antidepressant-like effects of ketamine in the forced swimming test. The phosphorylation of extracellular signal-regulated kinase 1/2 (pERK 1/2) was decreased by PD184161 in the amygdala and nucleus accumbens, and the effects of ketamine on pERK 1/2 in the prefrontal cortex and hippocampus were inhibited by PD184161. The ERK 2 levels were decreased by PD184161 in the nucleus accumbens; and the effects of ketamine were blocked in this brain area. The p38 protein kinase (p38MAPK) and proBDNF were inhibited by PD184161, and the MEK inhibitor prevented the effects of ketamine in the nucleus accumbens. In addition, ketamine increased pro-BDNF levels in the hippocampus. In conclusion, our findings demonstrated that an acute blockade of MAPK signaling lead to depressive-like behavior and stopped the antidepressant response of ketamine, suggesting that the effects of ketamine could be mediated, at least in part, by the regulation of MAPK signaling in these specific brain areas.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , Depressive Disorder/drug therapy , Ketamine/pharmacology , MAP Kinase Signaling System/drug effects , Aniline Compounds , Animals , Benzamides , Blotting, Western , Brain/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder/physiopathology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Neuropsychological Tests , Phosphorylation/drug effects , Rats, Wistar , Swimming , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Behav Brain Res ; 256: 451-6, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24004850

ABSTRACT

Studies indicate that histone deacetylation is important for long term changes related to stress and antidepressant treatment. The present study aimed to evaluate the effects of the classic antidepressant imipramine, and of an antagonist of the N-methyl-d-asparte (NMDA) receptor, ketamine, on behavior and histone deacetylase (HDAC) activity in the brains of maternally deprived adult rats. To this aim, deprived and non-deprived (control) male Wistar rats were divided into the following groups: non-deprived+saline; non-deprived+imipramine (30 mg/kg); non-deprived+ketamine (15 mg/kg); deprived+saline; deprived+imipramine (30 mg/kg); and deprived+ketamine (15 mg/kg). The drugs were administrated once a day for 14 days during their adult phase. Their behavior were then assessed using the forced swimming and open field tests. In addition, the HDAC activity was evaluated in the prefrontal cortex, hippocampus, amygdala and nucleus accumbens using the kit ELISA-sandwich test. In deprived rats treated with saline, we observed an increase in the immobility time, but treatments with imipramine and ketamine were able to reverse this alteration, decreasing the immobility time. Also, there was a decrease on number of crossings with imipramine treatment in non-deprived rats, and an increase on number of crossings with ketamine treatment in deprived rats. The HDAC activity did not alter in the prefrontal cortex, hippocampus and amygdala by deprivation or via treatment with imipramine or ketamine. However, in the nucleus accumbens we observed an increase of HDAC activity in the deprived rats, and interestingly, imipramine and ketamine treatments were able to decrease HDAC activity in this brain area. These findings provide a novel insight into the epigenetic regulation of histone deacetylase in the nucleus accumbens caused by imipramine and ketamine, and indicate that molecular events are necessary to reverse specific stress-induced behavior.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Histones/metabolism , Imipramine/pharmacology , Ketamine/pharmacology , Maternal Deprivation , Nucleus Accumbens/drug effects , Acetylation , Amygdala/drug effects , Amygdala/metabolism , Animals , Behavior, Animal/physiology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Swimming
SELECTION OF CITATIONS
SEARCH DETAIL
...