Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Psychiatr Res ; 175: 50-59, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38704981

ABSTRACT

Major depressive disorder (MDD) stands as a significant cause of disability globally. Cannabidiolic Acid-Methyl Ester (CBDA-ME) (EPM-301, HU-580), a derivative of Cannabidiol, demonstrates immediate antidepressant-like effects, yet it has undergone only minimal evaluation in psychopharmacology. Our goal was to investigate the behavioral and potential molecular mechanisms associated with the chronic oral administration of this compound in the Wistar Kyoto (WKY) genetic model of treatment-resistant depression. Male WKY rats were subjected to behavioral assessments before and after receiving chronic (14-day) oral doses of CBDA-ME (0.5 mg/kg), 15 mg/kg of imipramine or vehicle. At the end of the study, plasma corticosterone levels and mRNA expression of various genes in the medial Prefrontal Cortex and Hippocampus were measured. Behavioral outcomes from CBDA-ME treatment indicated an antidepressant-like effect similar to imipramine, as oral ingestion reduced immobility and increased swimming duration in the Forced Swim Test. Neither treatment influenced locomotion in the Open Field Test nor preference in the Saccharin Preference Test. The behavioral impact in WKY rats coincided with reduced corticosterone serum levels, upregulated mRNA expression of Cannabinoid receptor 1, Fatty Acid Amide Hydrolase, and Corticotropin-Releasing Hormone Receptor 1, alongside downregulation of the Serotonin Transporter in the hippocampus. Additionally, there was an upregulation of CB1 mRNA expression and downregulation of Brain-Derived Neurotrophic Factor in the mPFC. These findings contribute to our limited understanding of the antidepressant effects of CBDA-ME and shed light on its potential psychopharmacological mechanisms. This discovery opens up possibilities for utilizing cannabinoids in the treatment of major depressive disorder and related conditions.

2.
Int J Mol Sci ; 24(20)2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37894946

ABSTRACT

Depression is a serious neuropsychiatric disease affecting an increasing number of people worldwide. Cognitive deficits (including inattention, poor memory, and decision-making difficulties) are common in the clinical picture of depression. Cognitive impairment has been hypothesized to be one of the most important components of major depressive disorder (MDD; referred to as clinical depression), although typical cognitive symptoms are less frequent in people with depression than in people with schizophrenia or bipolar disorder (BD; sometimes referred to as manic-depressive disorder). The importance of α-Klotho in the aging process has been well-documented. Growing evidence points to the role of α-Klotho in regulating other biological functions, including responses to oxidative stress and the modulation of synaptic plasticity. It has been proven that a Klotho deficit may contribute to the development of various nervous system pathologies, such as behavioral disorders or neurodegeneration. Given the growing evidence of the role of α-Klotho in depression and cognitive impairment, it is assumed that this protein may be a molecular link between them. Here, we provide a research review of the role of α-Klotho in depression and cognitive impairment. Furthermore, we propose potential mechanisms (related to oxidative stress and glutamatergic transmission) that may be important in α-Klotho-mediated regulation of mental and cognitive function.


Subject(s)
Cognitive Dysfunction , Depressive Disorder, Major , Humans , Glucuronidase , Depression , Glutamic Acid , Synaptic Transmission
3.
Article in English | MEDLINE | ID: mdl-37088171

ABSTRACT

More than 80% of depressed patients struggle with learning new tasks, remembering positive events, or concentrating on a single topic. These neurocognitive deficits accompanying depression may be linked to functional and structural changes in the prefrontal cortex and hippocampus. However, their mechanisms are not yet completely understood. We conducted a narrative review of articles regarding animal studies to assess the state of knowledge. First, we argue the contribution of changes in neurotransmitters and hormone levels in the pathomechanism of cognitive dysfunction in animal depression models. Then, we used numerous neuroinflammation studies to explore its possible implication in cognitive decline. Encouragingly, we also observed a positive correlation between increased oxidative stress and a depressive-like state with concomitant memory deficits. Finally, we discuss the undeniable role of neurotrophin deficits in developing cognitive decline in animal models of depression. This review reveals the complexity of depression-related memory impairments and highlights the potential clinical importance of gathered findings for developing more reliable animal models and designing novel antidepressants with procognitive properties.


Subject(s)
Cognitive Dysfunction , Depression , Animals , Depression/complications , Depression/psychology , Rodentia , Cognitive Dysfunction/psychology , Antidepressive Agents , Memory Disorders/etiology , Disease Models, Animal
4.
Int J Mol Sci ; 24(4)2023 Feb 14.
Article in English | MEDLINE | ID: mdl-36835237

ABSTRACT

The pathophysiology of major depressive disorder (MDD) is diverse and multi-factorial, yet treatment strategies remain limited. While women are twice as likely to develop the disorder as men, many animal model studies of antidepressant response rely solely on male subjects. The endocannabinoid system has been linked to depression in clinical and pre-clinical studies. Cannabidiolic Acid-Methyl Ester (CBDA-ME, EPM-301) demonstrated anti-depressive-like effects in male rats. Here, we explored acute effects of CBDA-ME and some possible mediating mechanisms, using a depressive-like genetic animal model, the Wistar-Kyoto (WKY) rat. In Experiment 1, Female WKY rats underwent the Forced swim test (FST) following acute CBDA-ME oral ingestion (1/5/10 mg/kg). In Experiment 2, Male and female WKY rats underwent the FST after injection of CB1 (AM-251) and CB2 (AM-630) receptor antagonists 30 min before acute CBDA-ME ingestion (1 mg/kg, males; 5 mg/kg, females). Serum levels of Brain-Derived Neurotrophic Factor (BDNF), numerous endocannabinoids and hippocampal Fatty Acid Amide Hydrolase (FAAH) levels were assessed. Results indicate that females required higher doses of CBDA-ME (5 and 10 mg/kg) to induce an anti-depressive-like effect in the FST. AM-630 blocked the antidepressant-like effect in females, but not in males. The effect of CBDA-ME in females was accompanied by elevated serum BDNF and some endocannabinoids and low hippocampal expression of FAAH. This study shows a sexually diverse behavioral anti-depressive response to CBDA-ME and possible underlying mechanisms in females, supporting its potential use for treating MDD and related disorders.


Subject(s)
Cannabidiol , Depressive Disorder, Major , Receptor, Cannabinoid, CB2 , Animals , Female , Male , Rats , Brain-Derived Neurotrophic Factor , Cannabidiol/therapeutic use , Depressive Disorder, Major/drug therapy , Disease Models, Animal , Endocannabinoids , Rats, Inbred WKY , Receptor, Cannabinoid, CB2/antagonists & inhibitors
5.
Int J Mol Sci ; 23(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36232725

ABSTRACT

Depressive disorders (DDs) are an increasingly common health problem that affects all age groups. DDs pathogenesis is multifactorial. However, it was proven that stress is one of the most important environmental factors contributing to the development of these conditions. In recent years, there has been growing interest in the role of the glutamatergic system in the context of pharmacotherapy of DDs. Thus, it has become increasingly important to explore the functioning of excitatory synapses in pathogenesis and pharmacological treatment of psychiatric disorders (including DDs). This knowledge may lead to the description of new mechanisms of depression and indicate new potential targets for the pharmacotherapy of illness. An excitatory synapse is a highly complex and very dynamic structure, containing a vast number of proteins. This review aimed to discuss in detail the role of the key postsynaptic proteins (e.g., NMDAR, AMPAR, mGluR5, PSD-95, Homer, NOS etc.) in the excitatory synapse and to systematize the knowledge about changes that occur in the clinical course of depression and after antidepressant treatment. In addition, a discussion on the potential use of ligands and/or modulators of postsynaptic proteins at the excitatory synapse has been presented.


Subject(s)
Depressive Disorder , Synapses , Brain/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Disks Large Homolog 4 Protein/metabolism , Humans , Ligands , Synapses/metabolism
6.
Biol Psychiatry ; 90(2): 109-117, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34052037

ABSTRACT

The neurocircuitry that contributes to the pathophysiology of posttraumatic stress disorder and major depressive disorder, psychiatric conditions that exhibit a high degree of comorbidity, likely involves both overlapping and unique structural and functional changes within multiple limbic brain regions. In this review, we discuss neurobiological alterations that are associated with posttraumatic stress disorder and major depressive disorder and highlight both similarities and differences that may exist between these disorders to argue for the existence of a shared neurobiology. We highlight the key contributions based on preclinical studies, emerging from the late Professor Ronald Duman's research, that have shaped our understanding of the neurocircuitry that contributes to both the etiopathology and treatment of major depressive disorder and posttraumatic stress disorder.


Subject(s)
Depressive Disorder, Major , Stress Disorders, Post-Traumatic , Brain , Depression , Humans
7.
Int J Mol Sci ; 22(3)2021 Jan 21.
Article in English | MEDLINE | ID: mdl-33494322

ABSTRACT

Altered activity of fatty acid amide hydrolase (FAAH), an enzyme of the endocannabinoid system, has been implicated in several neuropsychiatric disorders, including major depressive disorder (MDD). It is speculated that increased brain FAAH expression is correlated with increased depressive symptoms. The aim of this scoping review was to establish the role of FAAH expression in animal models of depression to determine the translational potential of targeting FAAH in clinical studies. A literature search employing multiple databases was performed; all original articles that assessed FAAH expression in animal models of depression were considered. Of the 216 articles that were screened for eligibility, 24 articles met inclusion criteria and were included in this review. Three key findings emerged: (1) FAAH expression is significantly increased in depressive-like phenotypes; (2) genetic knockout or pharmacological inhibition of FAAH effectively reduces depressive-like behavior, with a dose-dependent effect; and (3) differences in FAAH expression in depressive-like phenotypes were largely localized to animal prefrontal cortex, hippocampus and striatum. We conclude, based on the animal literature, that a positive relationship can be established between brain FAAH level and expression of depressive symptoms. In summary, we suggest that FAAH is a tractable target for developing novel pharmacotherapies for MDD.


Subject(s)
Amidohydrolases/metabolism , Brain/metabolism , Brain/physiopathology , Depression/etiology , Depression/metabolism , Disease Susceptibility , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/genetics , Animals , Antidepressive Agents/pharmacology , Biomarkers , Brain/drug effects , Depression/drug therapy , Depression/psychology , Disease Models, Animal , Endocannabinoids/metabolism , Genetic Predisposition to Disease , Humans , Neurons/metabolism , Rodentia
8.
Eur J Neurosci ; 53(1): 114-125, 2021 01.
Article in English | MEDLINE | ID: mdl-31821620

ABSTRACT

Optimal acetylcholine (ACh) signaling is important for sustained attention and facilitates learning and memory. At the same time, human and animal studies have demonstrated increased levels of ACh in the brain during depressive episodes and increased symptoms of anxiety, depression, and reactivity to stress when ACh breakdown is impaired. While it is possible that the neuromodulatory roles of ACh in cognitive and affective processes are distinct, one possibility is that homeostatic levels of ACh signaling are necessary for appropriate learning, but overly high levels of cholinergic signaling promote encoding of stressful events, leading to the negative encoding bias that is a core symptom of depression. In this review, we outline this hypothesis and suggest potential neural pathways and underlying mechanisms that may support a role for ACh signaling in negative encoding bias.


Subject(s)
Acetylcholine , Memory , Animals , Brain , Humans , Learning , Neural Pathways
9.
Psychoneuroendocrinology ; 119: 104767, 2020 09.
Article in English | MEDLINE | ID: mdl-32563935

ABSTRACT

Major depressive disorder (MDD) is a common mental disorder characterized by a persistent feeling of sadness, slow thought, impaired focus and loss of interest but the underlying mechanisms are largely unknown. Dendritic spines play an important role in the formation and maintenance of emotional circuits in the brain. Abnormalities in this process can lead to psychiatric diseases. 7,8-Dihydroxy-4-methylcoumarin (Dhmc), a precursor in the synthesis of derivatives of 4-methyl coumarin, plays an important role in protecting the nervous system from developing diseases and its most distinctive feature is safety. The aim of this study was to investigate whether Dhmc alleviates chronic unpredictable mild stress (CUMS)-induced depression-like behaviors and reverses CUMS-induced alterations in dendritic spines of principal neurons in brain areas of the emotional circuits including the hippocampus, medial prefrontal cortex (mPFC), nucleus accumbens (NAc) and basolateral amygdala (BLA) in male rats. Our results showed that CUMS-induced depression-like behaviors were accompanied by a decrease in spine density in pyramidal neurons of both the hippocampal CA3 area and the mPFC, and an increase in spine density in both the neurons of BLA and the medium spiny neurons (MSNs) of the NAc, as well as a decrease in the levels of the AMPA receptor subunit GluA1 and Kalirin-7 in the hippocampus compared with the control group. Intraperitoneal injection (i.p.) of Dhmc to the CUMS-exposed rats ameliorated CUMS-induced depression-like behaviors and reversed CUMS-mediated alterations in spine density and the levels of both GluA1 and Kalirin-7. Our results show an important role of Dhmc in reversing CUMS-induced depression-like behaviors and CUMS-mediated alterations in spine density.


Subject(s)
Affect/drug effects , Coumarins/therapeutic use , Dendritic Spines/drug effects , Depression/drug therapy , Animals , Behavior, Animal/drug effects , Coumarins/pharmacology , Dendritic Spines/pathology , Dendritic Spines/physiology , Depression/pathology , Depression/psychology , Disease Models, Animal , Male , Nerve Net/drug effects , Nerve Net/pathology , Nerve Net/physiology , Rats , Rats, Sprague-Dawley , Stress, Psychological/drug therapy , Stress, Psychological/pathology , Stress, Psychological/physiopathology , Stress, Psychological/psychology
10.
Eur J Neurosci ; 52(8): 3979-3994, 2020 10.
Article in English | MEDLINE | ID: mdl-32320518

ABSTRACT

A significant number of adolescents are considered insufficiently active. This is of concern considering the negative association between physical activity and major depressive disorder (MDD). There is a lack of approved pharmacological treatment options in this population partly due to limited information on the risks associated with lasting effects during early life. Therefore, interest in non-pharmacological strategies is gaining popularity with low- to moderate-intensity exercise being especially attractive for its antidepressant-like effects and augmentation properties in combination with antidepressants. Early-life development might present a unique "window of opportunity" to induce long-term beneficial effects in individuals treated with central acting drugs, such as antidepressants. Therefore, we investigated the bio-behavioural effects of pre-pubertal, low-intensity exercise (EXE) and/or venlafaxine (VEN) on depressive-like behaviour in juvenile (postnatal day 35 (PND35)) and young adult (PND60) stress-sensitive Flinders sensitive line (FSL) rats. Interventions were introduced during pre-pubertal development, that is PND21-34, followed by a 26-day washout/sedentary period, when bio-behavioural analyses were performed in the early adulthood group. VEN, alone or in combination with EXE, proved ineffective in inducing any bio-behavioural changes in either age group. EXE did not induce early-life antidepressant-like effects, despite increasing frontal serotonin (5-HT) and noradrenaline (NA) levels. Later in life (PND60), pre-pubertal exercise reduced immobility and increased coping behaviours, together with increased cortical 5-HT levels, despite a significant reduction in locomotor activity. These findings emphasize a strong serotonergic basis to the observed delayed antidepressant effects of EXE later in life.


Subject(s)
Depressive Disorder, Major , Animals , Antidepressive Agents , Depression , Depressive Disorder, Major/drug therapy , Disease Models, Animal , Norepinephrine , Rats , Serotonin , Venlafaxine Hydrochloride/pharmacology
11.
BMJ Open Sci ; 3(1): e000017, 2019.
Article in English | MEDLINE | ID: mdl-35047681

ABSTRACT

INTRODUCTION: Major depressive disorder is the leading source of disability globally and current pharmacological treatments are less than adequate. Animal models such as the Flinders Sensitive Line (FSL) rats are used to mimic aspects of the phenotype in the human disorder and to characterise candidate antidepressant agents. Communication between the gut microbiome and the brain may play an important role in psychiatric disorders such as depression. Interventions targeting the gut microbiota may serve as potential treatments for depression, and this drives increasing research into the effect of probiotics and prebiotics in neuropsychiatric disorders. Prebiotics, galacto-oligosaccharides and fructooligosaccharides that stimulate the activity of gut bacteria have been reported to have a positive impact, reducing anxiety and depressive-like phenotypes and stress-related physiology in mice and rats, as well as in humans. Bimuno, the commercially available beta-galacto-oligosaccharide, has been shown to increase gut microbiota diversity. AIM: Here, we aim to investigate the effect of Bimuno on rat anxiety-like and depressive-like behaviour and gut microbiota composition in the FSL model, a genetic model of depression, in comparison to their control, the Flinders Resistant Line (FRL) rats. METHODS: Sixty-four male rats aged 5-7 weeks, 32 FSL and 32 FRL rats, will be randomised to receive Bimuno or control (4 g/kg) daily for 4 weeks. Animals will be tested by an experimenter unaware of group allocation on the forced swim test to assessed depressive-like behaviour, the elevated plus maze to assess anxiety-like behaviour and the open field test to assess locomotion. Animals will be weighed and food and water intake, per kilogram of bodyweight, will be recorded. Faeces will be collected from each animal prior to the start of the experiment and on the final day to assess the bacterial diversity and relative abundance of bacterial genera in the gut. All outcomes and statistical analysis will be carried out blinded to group allocation, group assignments will be revealed after raw data have been uploaded to Open Science Framework. Two-way analysis of variance will be carried out to investigate the effect of treatment (control or prebiotic) and strain (FSL or FRL) on depressive-like and anxiety-like behaviours.

12.
Physiol Behav ; 201: 59-63, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30571957

ABSTRACT

The current study explores the therapeutic potential of Cannabidiol (CBD), a compound in the Cannabis plant, using both sexes of 2 "depressive-like" genetic models, Wistar Kyoto (WKY) and Flinders Sensitive Line (FSL) rats. Rats ingested CBD (30 mg/kg) orally. In the saccharin preference test, following a previous report of a pro-hedonic effect of CBD in male WKY, we now found similar results in female WKY. CBD also decreased immobility in the forced swim test in males (both strains) and in female WKY. These findings suggest a role for CBD in treating mental disorders with prominent symptoms of helplessness and anhedonia.


Subject(s)
Antidepressive Agents/therapeutic use , Cannabidiol/therapeutic use , Depressive Disorder/drug therapy , Animals , Depressive Disorder/psychology , Disease Models, Animal , Female , Food Preferences/psychology , Male , Motor Activity/drug effects , Rats , Rats, Inbred WKY , Saccharin/pharmacology , Swimming/psychology
13.
Behav Brain Res ; 351: 1-3, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29860002

ABSTRACT

BACKGROUND AND PURPOSE: Cannabidiolic acid methyl ester (HU-580) was recently shown to reduce stress-induced anxiety-like behavior in rats. The aim of this study was to examine the antidepressant effect of HU-580 in two different rat models of depression. EXPERIMENTAL APPROACH: Using the forced swim test (FST), we evaluated the effect of HU-580 in 43 Wistar-Kyoto (WKY) and 23 Flinders Sensitive Line (FSL) adult male rats. KEY RESULTS: 1 mg/kg HU-580 reduced immobility and increased swimming in WKY rats, compared to vehicle-treated controls (p < 0.05). This dose exerted similar effects in FSL rats (p < 0.05). CONCLUSION AND IMPLICATIONS: This is the first report of antidepressant efficacy of HU-580. These findings expand the very limited existent results, suggesting that HU-580 is a potent anxiolytic agent. Taken together with its chemical stability, HU-580 emerges as a candidate for a future antidepressant medication.


Subject(s)
Antidepressive Agents/pharmacology , Cannabinoids/pharmacology , Depressive Disorder/drug therapy , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Motor Activity/drug effects , Rats, Inbred WKY , Species Specificity
14.
Behav Brain Res ; 351: 93-103, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29857028

ABSTRACT

A plethora of animal models of depression is described in the literature, aiming at mimicking different aspects of depression. Understanding the link between depression and stress has been and remains a major focus area for development of animal models, but lines of research with a more mechanistic focus targeting deficiencies in neurotransmitter systems or dysfunctional neuronal circuitries and neuroinflammation are also pursued vigorously. The main objectives of the present study were systematically to evaluate strain and sex characteristics of a genetic animal model, the Flinders Sensitive Line (FSL)/ Flinders Resistant Line (FRL), by applying behavioral, molecular and pharmacological measures relevant to depression, and compare it with the outbred Sprague Dawley rat. In addition, we aimed at comparing across strains and sex the expression of NPY, CRF, CGRP in brain regions critically involved in mood regulation, and investigating the responses to escitalopram. In line with the comparisons of FSL and FRL rats, the FSL rats weighed significantly less than SD rats. Overall, escitalopram treatment for 5-6 weeks did not have a major impact on weight, but displayed a significant antidepressant-like effect, however without any changes in NPY, CRH and CGRP expression. Our comparative study of FSL and SD rat with respect to behavioral characteristic, neuropeptide levels in various brain regions (protein and mRNA levels), and response to long-term antidepressant treatment revealed that female FSL rats showed the most pronounced depressive-like phenotype and response to SSRI treatment. However, these findings were not paralleled by changes in measures of NPY, CRH and CGRP function.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Citalopram/pharmacology , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Sex Characteristics , Animals , Brain/drug effects , Brain/physiopathology , Brain Edema , Disease Models, Animal , Female , Male , Neurodegenerative Diseases , Neuropeptides/metabolism , Optic Atrophy , Random Allocation , Rats , Spasms, Infantile , Species Specificity
15.
Brain Res ; 1652: 30-42, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27663969

ABSTRACT

Depression is one of the leading causes of disability and a significant health-concern worldwide. Much of our current understanding on the pathogenesis of depression and the pharmacology of antidepressant drugs is based on pre-clinical models. Three of the most popular stress-based rodent models are the forced swimming test, the chronic mild stress paradigm and the learned helplessness model. Despite their recognizable advantages and limitations, they are associated with an immense variability due to the high number of design parameters that define them. Only few studies have reported how minor modifications of these parameters affect the model phenotype. Thus, the existing variability in how these models are used has been a strong barrier for drug development as well as benchmark and evaluation of these pre-clinical models of depression. It also has been the source of confusing variability in the experimental outcomes between research groups using the same models. In this review, we summarize the known variability in the experimental protocols, identify the main and relevant parameters for each model and describe the variable values using characteristic examples. Our view of depression and our efforts to discover novel and effective antidepressants is largely based on our detailed knowledge of these testing paradigms, and requires a sound understanding around the importance of individual parameters to optimize and improve these pre-clinical models.


Subject(s)
Depressive Disorder , Disease Models, Animal , Stress, Psychological , Animals , Humans
16.
Ann N Y Acad Sci ; 1344: 37-49, 2015 May.
Article in English | MEDLINE | ID: mdl-25823952

ABSTRACT

Depression is a heterogeneous disorder and, similar to other spectrum disorders, its manifestation varies by age of onset, severity, comorbidity, treatment responsiveness, and other factors. A laboratory blood test based on specific biomarkers for major depressive disorder (MDD) and its subgroups could increase diagnostic accuracy and expedite the initiation of treatment. We identified candidate blood biomarkers by examining genome-wide expression differences in the blood of animal models representing both the genetic and environmental/stress etiologies of depression. Human orthologs of the resulting transcript panel were tested in pilot studies. Transcript abundance of 11 blood markers differentiated adolescent subjects with early-onset MDD from adolescents with no disorder (ND). A set of partly overlapping transcripts distinguished adolescent patients who had comorbid anxiety disorders from those with only MDD. In adults, blood levels of nine transcripts discerned subjects with MDD from ND controls. Even though cognitive behavioral therapy (CBT) resulted in remission of some patients, the levels of three transcripts consistently signaled prior MDD status. A coexpression network of transcripts seems to predict responsiveness to CBT. Thus, our approach can be developed into clinically valid diagnostic panels of blood transcripts for different manifestations of MDD, potentially reducing diagnostic heterogeneity and advancing individualized treatment strategies.


Subject(s)
Depressive Disorder, Major/blood , Gene Expression Profiling/methods , RNA/blood , Adolescent , Adult , Animals , Anxiety Disorders/blood , Anxiety Disorders/physiopathology , Anxiety Disorders/therapy , Biomarkers/blood , Cognitive Behavioral Therapy , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/therapy , Disease Models, Animal , Female , Humans , Pilot Projects
17.
J Neurochem ; 127(1): 114-23, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23600864

ABSTRACT

Neuropeptide galanin and its three G-protein coupled receptors, galanin receptor type 1-galanin receptor type 3 (GalR1-GalR3), are involved in the regulation of numerous physiological and disease processes, and thus represent tremendous potential in neuroscience research and novel drug lead development. One of the areas where galanin is involved is depression. Previous studies have suggested that activation of GalR2 leads to attenuation of depression-like behavior. Unfortunately, lack of in vivo usable subtype specific ligands hinders testing the role of galanin in depression mechanisms. In this article, we utilize an approach of increasing in vivo usability of peptide-based ligands, acting upon CNS. Thus, we have synthesized a series of novel systemically active galanin analogs, with modest preferential binding toward GalR2. We have shown that specific chemical modifications to the galanin backbone increase brain levels upon i.v. injection of the peptides. Several of the new peptides, similar to a common clinically used antidepressant medication imipramine, exerted antidepressant-like effect in forced swim test, a mouse model of depression, at a surprisingly low dose range (< 0.5 mg/kg). We chose one of the peptides, J18, for more thorough study, and showed its efficacy also in another mouse depression model (tail suspension test), and demonstrated that its antidepressant-like effect upon i.v. administration can be blocked by i.c.v. galanin receptor antagonist M35. The effect of the J18 was also abolished in GalR2KO animals. All this suggests that systemically administered peptide analog J18 exerts its biological effect through activation of GalR2 in the brain. The novel galanin analogs represent potential drug leads and a novel pharmaceutical intervention for depression.


Subject(s)
Behavior, Animal/drug effects , Depression/psychology , Receptor, Galanin, Type 2/drug effects , Amino Acid Sequence , Animals , Antidepressive Agents, Tricyclic/pharmacology , Binding, Competitive/drug effects , Cell Line, Tumor , Drug Design , Female , Galanin/metabolism , Hindlimb Suspension , Humans , Imipramine/pharmacology , Ligands , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/pharmacology , Swimming/psychology , Tissue Distribution
18.
Salud ment ; 35(5): 359-366, sep.-oct. 2012. ilus, mapas, tab
Article in Spanish | LILACS-Express | LILACS | ID: lil-675543

ABSTRACT

It has been proposed that gonadal hormones participate in regulation of mood and emotion in men as well as in the effect of psychoactive drugs, such as antidepressants. However, evaluation of this type of interactions has been poorly studied in clinic and basic studies. The objective of the present study was to determine the role of gonadal hormones, testosterone (T) and 17β-estradiol (E2), one of its main metabolites, in the effect of two antidepressant drugs: desipramine and fluoxetine. The former is a tricyclic antidepressant that inhibits noradrenaline reuptake in a preferential manner, while the second is a serotonin selective reup-take inhibitor (SSRI) and the most prescribed antidepressant. Behavioral evaluations were conducted in adult male rats, intact or orchidectomized (Orx), treated with T (0-2 mg/rata), E2 (0-40 µg/rata), desipramine (0-20 mg/kg), fluoxetine (0-20 mg/kg) and their combinations. Forced swimming test was used as an animal model to detect antidepressant-like effect induced by treatments, on the basis of its predictive validity. We found that desipramine and fluoxetine produced an anti-depressant-like effect in gonadally intact male rats. However, the antidepressant-like effect of both treatments was cancelled in Orx males. Treatment with E2, but not with T, produced antidepressant-like actions in Orx males. Interestingly, treatment with E2 restored the antidepressant-like effect of desipramine and fluoxetine, while supplementation with T only reestablished the antidepressant-like action of desipramine, evidencing that gonadal hormones have a differential participation in regulation of neurotransmitter systems involving in the antidepressant effect. In conclusion, the main testicular androgen T, participates in the expression of the effect of antidepressant drugs, mainly via conversion to its estrogenic metabolite E2. These results give support to the idea that a combined therapy of gonadal hormones and antidepressant drugs may be more convenient to treat depressive disorders in hypogonadal men resistant to conventional antidepressant drugs.


Se ha propuesto que las hormonas gonadales participan en la regulación del estado de ánimo en los varones, y en el efecto de los fármacos psicoactivos, tales como los antidepresivos. Sin embargo, la evaluación de este tipo de interacciones ha sido estudiada escasamente. El objetivo del presente trabajo fue determinar el papel que cumplen las hormonas testosterona (T) y 17β-estradiol (E2), uno de sus principales metabolitos, en el efecto de dos fármacos antidepresivos utilizados en la práctica clínica, desipramina y fluoxetina. El primero es un tricíclico con acciones sobre el sistema noradrenérgico, mientras que la fluoxetina es un inhibidor selectivo de la recaptura de serotonina. Las evaluaciones se llevaron a cabo utilizando ratas macho adultas jóvenes, gonadalmente intactas u orquidectomizadas (Orx), bajo tratamiento con T (0-1 mg/rata), E2 (0-40 µg/rata), desipramina (0-20 mg/kg), fluoxetina (0-20 mg/kg) y sus respectivas combinaciones. Se utilizó la prueba de nado forzado (PNF) para detectar las acciones antidepresivas de los tratamientos. Encontramos que desipramina y fluoxetina redujeron la conducta de depresión en los machos gonadalmente intactos; sin embargo, el efecto de ambos tratamientos fue abolido por la orquidectomía. El tratamiento de restitución hormonal con E2, pero no con T, indujo acciones antidepresivas en los machos Orx. A su vez, cuando los animales Orx recibieron la restitución con T se produjo la recuperación del efecto antidepresivo de la desipramina, mientras que el E2 restableció las acciones antidepresivas de ambos fármacos. En conclusión, el principal andrógeno de origen testicular, la T, participa en la expresión del efecto de los fármacos antidepresivos explorados en el presente estudio, principalmente a través de su metabolito estrogénico, el E2. Estos resultados apoyan la idea de que una terapia adjunta de tratamientos hormonales y antidepresivos sería de beneficio para varones hipogonadales que cursen con depresión resistente a los fármacos antidepresivos convencionales.

SELECTION OF CITATIONS
SEARCH DETAIL
...