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










Publication year range
1.
Eur Neuropsychopharmacol ; 43: 82-91, 2021 02.
Article in English | MEDLINE | ID: mdl-33341344

ABSTRACT

Higher risk of depression and schizophrenia in descendants of mothers experienced acute infection during the pregnancy has been reported. Since monoamines are fundamental in mentioned psychopathologies, it is possible that maternal immune activation leads to impaired functioning of serotonin (5-HT), noradrenaline, and dopamine neurons in offspring. To test this hypothesis, we examined the effect of maternal immune activation by lipopolysaccharide (LPS) in rats on the excitability of monoamine-secreting neurons in the offspring. LPS was administered during days 15-19 of the gestation in the rising doses of 20-80 µg/kg; control dams received vehicle. During days 53-63 postpartum, rats were anesthetized and electrodes were inserted into the dorsal raphe nucleus, locus coeruleus, and ventral tegmental area for in vivo excitability assessment of 5-HT, noradrenaline, and dopamine neurons. Maternal immune activation suppressed the firing rate of 5-HT neurons in both sexes and stimulated the firing rate of dopamine neurons in males. Decrease in the firing rate of 5-HT neurons was accompanied with an increase, and increase in the firing rate of dopamine neurons with a decrease, in the density of spontaneously active cells. Maternal immune activation also decreased the variability of interspike intervals in 5-HT and dopamine neurons. It is possible that the alteration of excitability of 5-HT and dopamine neurons by maternal immune activation is involved in the psychopathologies induced by infectious disease during the pregnancy. Stimulation of dopamine excitability in males might be a compensatory mechanism secondary to the maternal immune challenge-induced suppression of 5-HT neurons.


Subject(s)
Locus Coeruleus , Norepinephrine , Action Potentials , Animals , Dopaminergic Neurons , Female , Male , Pregnancy , Rats , Rats, Sprague-Dawley
2.
Behav Brain Res ; 398: 112944, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33017639

ABSTRACT

Preclinical studies suggest that stress-related disorders even prior gestation can cause long-term changes at the level of neurobehavioral adaptations. Therefore, it is critical to consider undergoing antidepressant therapy which could reverse the negative consequences in the offspring. Venlafaxine is widely used in clinical practice; however insufficient amount of well-controlled studies verified the safety of venlafaxine therapy during gestation and lactation. The aim of this work was to investigate the effects of perinatal venlafaxine therapy on selected neurobehavioral variables in mothers and their female offspring using a model of maternal adversity. Pre-gestational stressed and non-stressed Wistar rat dams were treated with either venlafaxine (10 mg/kg/day) or vehicle during pregnancy and lactation. We have shown that pre-gestational stress decreased the number of pups with a significant reduction in the number of males but not females. Furthermore, we found that offspring of stressed and treated mothers exhibited anxiogenic behavior in juvenile and adolescent age. However, during adulthood pre-gestational stress significantly increased anxiety-like behavior of female, with venlafaxine treatment normalizing the state to control levels. Additionally, we found that even maternal stress prior gestation can have long-term impact on adult number of hippocampal immature neurons of the female offspring. A number of questions related to the best treatment options for maternal depression still remains, however present data may provide greater insight into the possible outcomes associated with perinatal venlafaxine therapy.


Subject(s)
Anxiety/etiology , Hippocampus/growth & development , Maternal Behavior/drug effects , Maze Learning/drug effects , Prenatal Exposure Delayed Effects/etiology , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Stress, Psychological/drug therapy , Venlafaxine Hydrochloride/pharmacology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Female , Lactation , Postpartum Period , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Rats , Rats, Wistar , Serotonin and Noradrenaline Reuptake Inhibitors/administration & dosage , Sex Factors , Venlafaxine Hydrochloride/administration & dosage
3.
ASN Neuro ; 12: 1759091420925335, 2020.
Article in English | MEDLINE | ID: mdl-32443939

ABSTRACT

Microglia, the resident immune cells of the central nervous system, are not a homogeneous population; their morphology, molecular profile, and even their ultrastructure greatly vary from one cell to another. Recent advances in the field of neuroimmunology have helped to demystify the enigma that currently surrounds microglial heterogeneity. Indeed, numerous microglial subtypes have been discovered such as the disease-associated microglia, neurodegenerative phenotype, and Cd11c-positive developmental population. Another subtype is the dark microglia (DM), a population defined by its ultrastructural changes associated with cellular stress. Since their first characterization using transmission electron microscopy, they have been identified in numerous disease conditions, from mouse models of Alzheimer's disease, schizophrenia, fractalkine signaling deficiency to chronic stress, just to name a few. A recent study also identified the presence of cells with a similar ultrastructure to the DM in postmortem brain samples from schizophrenic patients, underlining the importance of understanding the function of these cells. In this minireview, we aim to summarize the current knowledge on the DM, from their initial ultrastructural characterization to their documentation in various pathological contexts across multiple species. We will also highlight the current limitations surrounding the study of these cells and the future that awaits the DM.


Subject(s)
Brain/pathology , Brain/ultrastructure , Microglia/pathology , Microglia/ultrastructure , Microscopy, Electron, Transmission/methods , Animals , Brain/metabolism , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/ultrastructure , Humans , Mental Disorders/metabolism , Mental Disorders/pathology , Microglia/metabolism , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology
4.
Molecules ; 24(23)2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31757051

ABSTRACT

SMe1EC2M3 is a pyridoindole derivative related to the neuroleptic drug carbidine. Based on the structural similarities of SMe1EC2M3 and known serotonin (5-HT), norepinephrine, and dopamine reuptake inhibitors, we hypothesized that this compound may also have triple reuptake inhibition efficacy and an antidepressant-like effect. PreADMET and Dragon software was used for in silico prediction of pharmacokinetics and pharmacodynamics of SMe1EC2M3. Forced swim test was used to evaluate its antidepressant-like effects. Extracellular in vivo electrophysiology was used to assess 5-HT, norepinephrine, and dopamine reuptake inhibition efficacy of SMe1EC2M3. PreADMET predicted reasonable intestinal absorption, plasma protein binding, and blood-brain permeability for SMe1EC2M3. Dragon forecasted its efficiency as an antidepressant. Using behavioral measurements, it was found that SMe1EC2M3 decreased immobility time and increase swimming time during the forced swim test (FST). Electrophysiological investigations showed that SMe1EC2M3 dose-dependently suppressed the excitability of 5-HT neurons of the dorsal raphe nucleus (DRN), norepinephrine neurons of the locus coeruleus (LC), and dopamine neurons of the ventral tegmental area (VTA). The SMe1EC2M3-induced suppression of 5-HT, norepinephrine, and dopamine neurons was reversed by the antagonists of serotonin-1A (5-HT1A; WAY100135), α-2 adrenergic (α2, yohimbine), and dopamine-2 receptors (D2, haloperidol), respectively. We conclude that SMe1EC2M3 is prospective triple 5-HT, norepinephrine, and dopamine reuptake inhibitor with antidepressant-like properties, however future studies should be performed to complete the pharmacological profiling of this compound.


Subject(s)
Antidepressive Agents , Electrophysiological Phenomena/drug effects , Neurons/metabolism , Neurotransmitter Uptake Inhibitors , Synaptic Transmission/drug effects , Animals , Antidepressive Agents/chemistry , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/pharmacology , Male , Neurotransmitter Uptake Inhibitors/chemistry , Neurotransmitter Uptake Inhibitors/pharmacokinetics , Neurotransmitter Uptake Inhibitors/pharmacology , Rats , Rats, Wistar
5.
Behav Brain Res ; 375: 112131, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31377253

ABSTRACT

Chronic stress during pregnancy or even prior to gestation can negatively affect offspring´s neurobehavioural development. Several studies have shown, that offspring who had experienced excessive stress during gestation had higher rates of cognitive and mood disorders later during adolescence or in adulthood. Hippocampal neurons play a crucial role in the regulation of behavior, mainly in anxiety-related behaviors and spatial learning and memory. Recently, it has been shown, that excessive stress even prior to gestation could interfere with sensitive developmental processes in the brain and may affect hippocampal functioning with severe neurobehavioural consequences in later life. The aim of this work was to investigate the effects of pre-gestational stress of the rat dams on the hippocampal excitability of the pups right after the birth. Neurobehavioural consequences of pre-gestational stress were analyzed during adolescence (35-40 postnatal days) and in early adulthood (75-80 postnatal days). We have shown that even pre-gestational chronic maternal stress increased resting membrane potential, suppressed depolarization-activated action potential firing, and increased spontaneous activity of hippocampal cells from newborn offspring. Altered function of hippocampus was reflected at the behavioural level. Adolescent male offspring of dams exposed stress prior to conception showed hyperactivity-like behaviour in a new stressful environment and increased anxiety-like behaviour during adulthood compared to adult males from non-stress group. Together, this work suggests, that chronic stress even prior to gestation can interfere with functional brain development of the offspring and can cause long-term behavioural changes at the level of neurobehavioural adaptations.


Subject(s)
Hippocampus/pathology , Pregnancy Complications/pathology , Pregnancy Complications/psychology , Stress, Psychological/pathology , Stress, Psychological/psychology , Action Potentials/physiology , Animals , Anxiety/etiology , Anxiety/psychology , Attention Deficit Disorder with Hyperactivity/psychology , Cells, Cultured , Chronic Disease , Feeding Behavior , Female , Male , Maze Learning , Membrane Potentials/physiology , Neurogenesis/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Wistar
6.
Biomed Res Int ; 2019: 6149023, 2019.
Article in English | MEDLINE | ID: mdl-31976324

ABSTRACT

The aim of this study was to translate the "orientation" query of the ADAS-Cog inventory to rats and to investigate whether they can determine which time of the day they are. For this purpose, we established a modified Morris water-maze navigation task where the escape platform was placed onto various locations at different times of the day: "morning", "noon" and "evening". In each of these sessions rats swam a "query" trial and a "confirmatory" trial, 30 min apart. Lister Hooded rats randomly chose among the three possible target locations, while Long Evans rats partly followed a win-stay strategy by preferring to visit first to the platform position of the previous session. Despite simplifying the task to a morning-evening discrimination, Lister Hooded rats continued searching by chance, while Long Evans rats switched to the mentally less demanding random strategy. We then inserted a board into the pool which required longer swimming path from the animals when they were correcting an initial wrong choice, but this modification did not result in a change in the above strategies. Lastly, in a separate group of Long-Evans rats, the training conditions were modified inasmuch an incorrect choice was definitely punished by impeding the animals to correct it and confining them to a platform-free part of the maze for the whole trial period. However, even these stricter conditions were not sufficient to make the rats distinguish times of the day. The observed lack of time discrimination may source from an evolutionary built in mechanism characteristic for the rat species or this ability may have only been lost in laboratory rats.


Subject(s)
Discrimination Learning/physiology , Maze Learning/physiology , Orientation/physiology , Rats/psychology , Alzheimer Disease , Animals , Male , Rats, Long-Evans , Reaction Time/physiology , Swimming
7.
Behav Pharmacol ; 29(5): 445-452, 2018 08.
Article in English | MEDLINE | ID: mdl-29561291

ABSTRACT

Depression during pregnancy and in the post-partum period is a growing health issue. Venlafaxine, a representative of serotonin and noradrenaline reuptake inhibitors, is used to treat a wide spectrum of mood disorders. However, the limited number of prenatal and perinatal studies raises the question about the long-term consequences of venlafaxine therapy. The aim of this study was to investigate the effect of venlafaxine exposure during pregnancy and lactation on anxiety-like and depression-like behaviors, as well as adrenocortical hormone concentrations in the adult rat offspring. For this purpose, rat dams were treated orally with venlafaxine from day 15 of gestation to postnatal day 20 at doses of 7.5, 37.5, and 75 mg/kg. Administration of venlafaxine during gestation and lactation affected anxiety-like and depression-like behaviors in adult rat offspring of both sexes. The animals exposed through their mothers to venlafaxine, particularly at the lowest and middle doses, were less anxious and less depressive in several relevant behavioral tests, which can be considered a deviation from the normal state. At clinically relevant doses, venlafaxine did not alter circulating level of corticosterone and aldosterone in the adult offspring. In general, the consequences of venlafaxine were dose dependent and more apparent in females. Together, these results suggest that prenatal and early postnatal exposure to venlafaxine may interfere with functional development of the brain, though not necessarily in a negative way.


Subject(s)
Anxiety/drug therapy , Postpartum Period/drug effects , Venlafaxine Hydrochloride/pharmacology , Adrenal Cortex Hormones/analysis , Adrenal Cortex Hormones/blood , Aldosterone , Animals , Animals, Newborn/metabolism , Anxiety/metabolism , Anxiety Disorders/drug therapy , Behavior, Animal/drug effects , Brain/drug effects , Corticosterone , Depression/drug therapy , Depressive Disorder/physiopathology , Female , Fluoxetine/pharmacology , Hippocampus/drug effects , Male , Maternal Behavior/drug effects , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/physiopathology , Venlafaxine Hydrochloride/metabolism
8.
Neurosci Biobehav Rev ; 85: 102-116, 2018 02.
Article in English | MEDLINE | ID: mdl-28472631

ABSTRACT

Recent research has linked early life exposure to selective serotonin reuptake inhibitor medications (SSRIs) to modifications of social behaviors in children. Serotonin is a key regulator of neurodevelopment, social behaviors and mental health, and with the growing use of SSRIs to treat maternal affective disorders during the perinatal period, questions have been raised about the benefits and risks of perinatal SSRI exposure on the developing child. This review will highlight how perinatal SSRIs affect maternal care and neurodevelopmental outcomes related to social affiliative behaviors in offspring; such as play behaviors, social interactions, reproductive behaviors, and maternal care of the next generation. We will also review how early life exposure to SSRIs can alter related neurobiology, and the epigenome. Both clinical research and findings from animal models will be discussed. Understanding the impact of perinatal SSRIs on neurobehavioral outcomes will improve the health and well-being of subsequent generations.


Subject(s)
Mood Disorders/drug therapy , Prenatal Exposure Delayed Effects/drug therapy , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/pharmacology , Social Behavior , Animals , Depression/drug therapy , Female , Humans , Pregnancy
9.
Behav Brain Res ; 339: 222-231, 2018 Feb 26.
Article in English | MEDLINE | ID: mdl-29203333

ABSTRACT

There is increasing evidence that mental health concerns, stress-related mental illnesses, and parental stress prior to conception have long-term effects on offspring outcomes. However, more work is needed to understand how pre-gestational stress might affect neurobehavioral outcomes in the mother. We investigated how chronic stress prior to gestation affects maternal behavior and related physiology, and aimed to determine the role that perinatal SSRIs have in altering these stress effects. To do this, female Sprague-Dawley rats were subject to chronic unpredictable stress (CUS) prior to breeding. During the perinatal period they were administered fluoxetine (10mg/kg/day). Four groups of dams were studied: Control+Vehicle, Pre-gestational Stress+Vehicle, Control+Fluoxetine and Pre-gestational Stress+Fluoxetine. Maternal weight, breeding success, and maternal caregiving behaviors were recorded. Measures of serum corticosterone and corticosteroid-binging globulin (CBG) and the number of immature neurons in the dorsal hippocampus were also assessed in the late postpartum. Main findings show pre-gestational stress resulted in poor reproductive success and maintenance of pregnancy. Pre-gestationally stressed dams also showed higher levels of nursing and fewer bouts of licking/grooming offspring in the first week postpartum - behaviors that were not reversed by perinatal fluoxetine treatment. In the dam, perinatal fluoxetine treatment reversed the effect of pre-gestational maternal stress on serum corticosterone levels and increased serum CBG levels as well as neurogenesis in the dorsal hippocampus. Maternal corticosterone levels significantly correlated with blanket and passive nursing. This work provides evidence for a long-term impact of stress prior to gestation in the mother, and shows that perinatal SSRI medications can prevent some of these effects.


Subject(s)
Behavior, Animal/drug effects , Corticosterone/blood , Fluoxetine/pharmacology , Maternal Behavior/drug effects , Animals , Female , Hippocampus/drug effects , Neurogenesis/drug effects , Postpartum Period/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/drug therapy , Rats, Sprague-Dawley , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Psychological/drug therapy
10.
Psychoneuroendocrinology ; 84: 159-171, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28735226

ABSTRACT

Selective serotonin reuptake inhibitor medications (SSRIs) are the first lines of treatment for maternal affective disorders, and are prescribed to up to 10% of pregnant women. Concern has been raised about how perinatal exposure to these medications affect offspring neurobehavioral outcomes, particularly those related to social interactions, as recent research has reported conflicting results related to autism spectrum disorder (ASD) risk in children prenatally exposed to SSRIs. Therefore, the aim of this work was to investigate the effects of perinatal exposure to the SSRI fluoxetine on social play behaviors and the hypothalamic pituitary adrenal system, using a model of pre-gestational maternal stress. We also investigated synaptic proteins in the CA2, CA3, and dentate gyrus of the hippocampus, as well as number of immature neurons in the granule cell layer, as both measures of plasticity in the hippocampus have been linked to social behaviors. In pre-adolescent male and female Sprague-Dawley rat offspring, main findings show that perinatal fluoxetine prevents the negative effect of maternal stress on sibling play behavior. However, perinatal fluoxetine increased social aggressive play with a novel conspecific in both sexes and decreased time grooming a novel conspecific in males only. Perinatal fluoxetine also increased serum corticosteroid binding globulin levels, 5-HT levels in the hippocampus, and pre-synaptic density assessed via synaptophysin in the dentate gyrus. Social interaction was significantly correlated with changes in plasticity in the CA2 region of the hippocampus. Pre-gestational maternal stress exposure resulted in significantly decreased rates of hippocampal neurogenesis and synaptophysin density in the dentate gyrus of pre-adolescent males, but not females. Together, these results further characterize the role of perinatal SSRIs, maternal stress prior to conception, and sex/gender on developing social behaviors and related plasticity in the hippocampus of pre-adolescent offspring.


Subject(s)
Fluoxetine/adverse effects , Stress, Psychological/metabolism , Animals , Anxiety/drug therapy , Behavior, Animal/drug effects , Depression/drug therapy , Female , Fluoxetine/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Interpersonal Relations , Male , Maternal Exposure , Neurogenesis/drug effects , Neuronal Plasticity/drug effects , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley , Selective Serotonin Reuptake Inhibitors/pharmacology , Sex Factors
11.
Neuro Endocrinol Lett ; 38(1): 19-26, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28456144

ABSTRACT

OBJECTIVE: Epidemiological studies strongly support the theory that stressful life events play an important role in the etiology of depression. The mechanism of chronic stress induced depression involves a number of systems. Chronic stress represents a serious health issue especially during pregnancy and lactation. In this sensitive period, stress can lead to changes in emotion and cognitive behavior both of the mothers and the offspring. It is thus necessary to properly manage stress events during gestation. Venlafaxine belongs to the group of serotonin and noradrenaline re-uptake inhibitor drugs. It is used for the treatment of depression, anxiety disorders and other mood disorders. During pregnancy, however, the use of venlafaxine is questionable due to the lack of experimental and clinical studies. Therefore the aim of this study was to evaluate the effect of chronic unpredictable stress and/or venlafaxine treatment on maternal and open field behavior of dams. Moreover, hippocampal neurogenesis was investigated either. METHODS: Female Wistar rats were subjected to 2-week chronic unpredictable stress induced by random stressors and treated with venlafaxine orally at a dose of 5 mg/kg twice a day. Maternal behavior was evaluated within 5-min observations twice a day. Mothers were also tested in the open field 8 weeks after chronic unpredictable stress procedure in a single 15-min session. Hippocampal neurogenesis was investigated by immunohistochemistry essay using DCX staining. RESULTS: Results of the present study showed altered maternal and open field behavior of the dams. Stressed dams had lowered hippocampal neurogenesis, while venlafaxine treatment reversed this lowering. CONCLUSIONS: These results suggest that stress and antidepressant therapy can have significant impact on behavior and hippocampal neurogenesis in rat dams.


Subject(s)
Behavior, Animal/drug effects , Hippocampus/drug effects , Maternal Behavior/drug effects , Neurogenesis/drug effects , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Stress, Psychological/psychology , Venlafaxine Hydrochloride/pharmacology , Animals , Disease Models, Animal , Doublecortin Protein , Female , Hippocampus/cytology , Hippocampus/metabolism , Immunohistochemistry , Pregnancy , Rats , Rats, Wistar
12.
Interdiscip Toxicol ; 10(1): 30-34, 2017 Sep.
Article in English | MEDLINE | ID: mdl-30123033

ABSTRACT

At present, affective disorders are among the most commonly diagnosed mental diseases. In pregnancy, they can occur as pre-delivery depression, recurrent depressive disorder or postnatal depression. The estimated prevalence of depressive disorders in pregnancy is approximately 9-16%, with some statistics reporting up to 20%. Approximately 2-3% of pregnant women take antidepressants during pregnancy, and the number of mothers treated increases by birth to 5-7%. Treatment of depression during pregnancy and breastfeeding is a controversial issue, as antidepressants can negatively affect the developing fetus. According to epidemiological studies, the effects of treated depression in pregnancy are related to premature birth, decreased body weight of the child, intrauterine growth retardation, neonatal adaptive syndrome, and persistent pulmonary hypertension. However, untreated depression can adversely affect maternal health and increase the risk of preeclampsia and eclampsia, as well as of subsequent postnatal depression, which can lead to disruption of the mother-child relationship. Based on the above mentioned facts, the basic question arises as to whether or not to treat depression during pregnancy and lactation.

13.
Interdiscip Toxicol ; 10(1): 35-39, 2017 Sep.
Article in English | MEDLINE | ID: mdl-30123034

ABSTRACT

Depression is one of the most prevalent and life-threatening forms of mental illness affecting about 20% of the population. Depressive disorder as a biochemical phenomenon, was first recognized in the mid-20th century of research, however the etiology of this disease is still not well understood. Although the need to investigate depressive disorders has emerged from the needs of clinical practice, there are many preclinical studies, which brought new insights into this field of research. During experimental work it was crucial to develop appropriate animal models, where the neurohumoral mechanism was similar to humans. In the past decades, several animal models of maternal depression have been developed. We describe the three most popular rodent models of maternal depression which are based on 1. stress prior to gestation, 2. prenatal stress and 3. early life stress. The above-mentioned animal models appear to fulfill many criteria for a relevant animal model of depression; they alter the regulation of the HPA, induce signs of depression-like behavior and several antidepressant treatments can reverse the state induced by maternal stress. Although, they are not able to model all aspects of maternal depression, they are useful models for monitoring neurodevelopmental changes occurring in dams and offspring.

14.
Interdiscip Toxicol ; 10(1): 40-43, 2017 Sep.
Article in English | MEDLINE | ID: mdl-30123035

ABSTRACT

An animal model of human behavior represents a complex of cognitive and/or emotional processess, which are translated from animals to humans. A behavioral test is developed primarily and specifically to verify and support a theory of cognition or emotion; it can also be used to verify a theory of a psychopathology, but it is not developed for a particular type of psychopathology. The paper reviews tests commonly used in novel drug discovery research. Focus is especially on tests which can evaluate anxiety-like (openfield test, novelty suppressed feeding, elevated plus maze, light/dark box, stressinduced hyperthermia) and depression-like behaviors (forced swim test, tail suspension test, sucrose preference test) as they represent an important methodological tool in pre-clinical as well as in behavioral toxicology studies.

15.
Neuro Endocrinol Lett ; 37(Suppl1): 103-110, 2016 Dec 18.
Article in English | MEDLINE | ID: mdl-28263537

ABSTRACT

OBJECTIVES: There are several models of depression. Chronic unpredictable mild stress (CMS) appears to have the greatest validity, although it is often being criticized for low reliability. METHODS: Male Wistar/DV rats were used in this study to assess our modified 2-week model of CMS as a combination of psychosocial, physical and metabolic stressors and to compare the effect of acute administration of venlafaxine (VFX) and diazepam (DZP), either in stress or no stress conditions. The animals were exposed to one particular stressor each day. The time of day and duration of the stressor differed across the procedure to avoid animals to adapt to the stress stimulus. After cessation of stress, the animals underwent the following behavioral tests to assess motor activity, cognition, anxiety- and depression-like behavior: Open field test, Elevated plus maze, Forced swim test, Stress-iduced hyperthermia, Light/dark test and Y maze. To assess hypothalamic-pituitary-adrenal axis (HPA) reactivity in our CMS model, plasma corticosterone levels were measured 24 h after termination of stress. RESULTS: Corticosterone levels were significantly increased compared to control values (p<0.05) in our experimental schedule of CMS. Our paradigm produced delayed anxiety-like behavior observed in Open field (decreased time spent in central zone 3 weeks after CMS, p<0.05), with anxiolytic effect of CMS shortly after its cessation. Stressed animals spent more time in the open arms of Elevated plus maze (p<0.05) and travelled longer distance in the light zone of the Light/dark box (p<0.01). CMS did not increase the behavioral despair analyzed in Forced swim test yet it disrupted the capacity of the Stress-induced hyperthermia test (CMS rats failed to react to the stress by increasing the core temperature). CONCLUSIONS: Based on our results, we can conclude that our CMS protocol leads to increased corticosterone levels as a result of HPA axis hyperactivity and produces delayed onset of anxiogenic behavior. Moreover, CMS exerted a substantial effect on the behavioral outputs, interfering with drug testing.


Subject(s)
Anxiety/physiopathology , Behavior, Animal/physiology , Corticosterone/blood , Depression/physiopathology , Stress, Psychological/physiopathology , Animals , Disease Models, Animal , Male , Rats , Rats, Wistar , Stress, Psychological/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...