Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 175
Filter
1.
Neuropsychopharmacology ; 46(6): 1161-1171, 2021 05.
Article in English | MEDLINE | ID: mdl-33564104

ABSTRACT

Cognitive dysfunction in schizophrenia (SZ) is thought to arise from neurodevelopmental abnormalities that include interneuron hypomyelination in the prefrontal cortex (PFC). Here we report that RNA-sequencing of the medial (m)PFC of the APO-SUS rat model with SZ-relevant cognitive inflexibility revealed antioxidant metabolism as the most-enriched differentially expressed pathway. Antioxidant-related gene expression was altered throughout postnatal development and preceded hypomyelination. Furthermore, reduced glutathione levels and increased mitochondria numbers were observed in the mPFC. Strikingly, chronic treatment with the glutathione precursor N-acetylcysteine (NAC) from postnatal days 5-90 restored not only antioxidant-related mRNA expression and mitochondria numbers, but also myelin-related mRNA expression and mPFC-dependent cognitive dysfunction, while blood glutathione levels remained unaffected. The promyelinating effect of NAC was at least partly due to a positive effect on oligodendrocyte lineage progression. Together, our findings highlight that oxidative stress may contribute to cognitive symptoms in the APO-SUS rat model of SZ and encourage antioxidant therapy in early phases of SZ.


Subject(s)
Cognitive Dysfunction , Schizophrenia , Animals , Antioxidants/pharmacology , Cognition , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Prefrontal Cortex , Rats , Schizophrenia/complications , Schizophrenia/drug therapy
2.
Biochem Pharmacol ; 178: 114050, 2020 08.
Article in English | MEDLINE | ID: mdl-32446887

ABSTRACT

Efavirenz (EFV) is used for antiretroviral treatment of HIV infection, and successfully inhibits viral replication and mother-to-child transmission of HIV during pregnancy and childbirth. Unfortunately, the drug induces neuropsychiatric symptoms such as anxiety and depressed mood and potentially affects cognitive performance. EFV acts on, among others, the serotonin transporter and serotonin receptors that are expressed in the developing brain. Yet, how perinatal EFV exposure affects brain cytoarchitecture remains unclear. Here, we exposed pregnant and lactating rats to EFV, and examined in the medial prefrontal cortex (mPFC) of their adult offspring the effects of the maternal EFV exposure on cortical architecture. We observed a significant decrease in the number of cells, mainly mature neurons, in the infra/prelimbic and cingulate cortices of adult offspring. Next, we found an altered cortical cytoarchitecture characterized by a significant reduction in deep- and superficial-layer cells. This was accompanied by a sharp increase in programmed cell death, as we identified a significantly higher number of cleaved Caspase-3-positive cells. Finally, the serotonergic and dopaminergic innervation of the mPFC subdomains was increased. Thus, the perinatal exposure to EFV provoked in the mPFC of adult offspring cell death, significant changes in cytoarchitecture, and disturbances in serotonergic and dopaminergic innervation. Our results are important in the light of EFV treatment of HIV-positive pregnant women, and its effect on brain development and cognitive behavior.


Subject(s)
Alkynes/toxicity , Benzoxazines/toxicity , Cyclopropanes/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/pathology , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Reverse Transcriptase Inhibitors/toxicity , Animals , Animals, Newborn , Anti-HIV Agents/toxicity , Female , Male , Prefrontal Cortex/growth & development , Pregnancy , Rats , Rats, Wistar
3.
Neurosci Biobehav Rev ; 114: 53-69, 2020 07.
Article in English | MEDLINE | ID: mdl-32320813

ABSTRACT

In the Western world, 2-5 % of pregnant women use selective serotonin reuptake inhibitor (SSRI) antidepressants. There is no consensus on the potential long-term neurodevelopmental outcomes of early SSRI exposure. Our aim was to determine whether there is an overall effect of perinatal SSRI exposure in animals on a spectrum of behavioral domains. After a comprehensive database search in PubMed, PsycINFO, and Web of Science, we included 99 publications. We performed nine meta-analyses and two qualitative syntheses corresponding to different behavioral categories, aggregating data from thousands of animals. We found evidence for reduced activity and exploration behavior (standardized mean difference (SMD) -0.28 [-0.38, -0.18]), more passive stress coping (SMD -0.37 [-0.52, -0.23]), and less efficient sensory processing (SMD -0.37 [-0.69, -0.06]) in SSRI- versus vehicle-exposed animals. No differences were found for anxiety (p = 0.06), social behavior, learning and memory, ingestive- and reward behavior, motoric behavior, or reflex and pain sensitivity. Exposure in the period equivalent to the human third trimester was associated with the strongest effects.


Subject(s)
Prenatal Exposure Delayed Effects , Selective Serotonin Reuptake Inhibitors , Animals , Antidepressive Agents , Anxiety , Female , Humans , Pregnancy , Social Behavior
4.
Neurosci Biobehav Rev ; 92: 338-349, 2018 09.
Article in English | MEDLINE | ID: mdl-29940236

ABSTRACT

The biological background and consequences of serotonin transporter polymorphism-glucocorticoid relationship in individual differences in stress reactivity has been a major interest in neuropsychiatry research. Individual differences in glucocorticoid release have long been implicated in vulnerability to stress-related psychopathologies, like depression and anxiety in various species. Yet, it is largely elusive to what extent results from non-human primates and rodents translate to human findings. Based on our structured, comprehensive and non-hypothesis driven overview of this topic, we conclude that although gene-environment interaction studies have highlighted the importance of serotonin transporter polymorphism in modulating glucocorticoid release, there is compelling evidence that age, gender and ethnicity are significant factors too contributing to the equation. We conclude too that the way early life events modulate an individual's stress reactivity as a function of serotonin transporter polymorphism is comparable between species. These data provide a rationale for the design of new, prospective translational studies into sex-specific gene-environment interactions across the lifespan with the goal of improving preventative efforts and optimizing (personalized) treatment in stress-related psychopathologies.


Subject(s)
Glucocorticoids/metabolism , Polymorphism, Genetic/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Stress, Psychological/genetics , Stress, Psychological/metabolism , Animals , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiopathology
5.
Drug Alcohol Depend ; 187: 186-194, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29679913

ABSTRACT

BACKGROUND: Neuroimaging studies have demonstrated gray matter (GM) volume abnormalities in substance users. While the majority of substance users are polysubstance users, very little is known about the relation between GM volume abnormalities and polysubstance use. METHODS: In this study we assessed the relation between GM volume, and the use of alcohol, tobacco, cocaine and cannabis as well as the total number of substances used, in a sample of 169 males: 15 non-substance users, 89 moderate drinkers, 27 moderate drinkers who also smoke tobacco, 13 moderate drinkers who also smoke tobacco and use cocaine, 10 heavy drinkers who smoke tobacco and use cocaine and 15 heavy drinkers who smoke tobacco, cannabis and use cocaine. RESULTS: Regression analyses showed that there was a negative relation between the number of substances used and volume of the dorsal medial prefrontal cortex (mPFC) and the ventral mPFC. Without controlling for the use of other substances, the volume of the dorsal mPFC was negatively associated with the use of alcohol, tobacco, and cocaine. After controlling for the use of other substances, a negative relation was found between tobacco and cocaine and volume of the thalami and ventrolateral PFC, respectively. CONCLUSION: These findings indicate that mPFC alterations may not be substance-specific, but rather related to the number of substances used, whereas, thalamic and ventrolateral PFC pathology is specifically associated with tobacco and cocaine use, respectively. These findings are important, as the differential alterations in GM volume may underlie different cognitive deficits associated with substance use disorders.


Subject(s)
Alcoholism/diagnostic imaging , Cocaine-Related Disorders/diagnostic imaging , Gray Matter/diagnostic imaging , Marijuana Abuse/diagnostic imaging , Self Report , Tobacco Use Disorder/diagnostic imaging , Adolescent , Adult , Alcohol Drinking/adverse effects , Alcohol Drinking/epidemiology , Alcohol Drinking/trends , Alcoholism/epidemiology , Cannabis/adverse effects , Cocaine/administration & dosage , Cocaine/adverse effects , Cocaine-Related Disorders/epidemiology , Drug Users , Ethanol/administration & dosage , Ethanol/adverse effects , Gray Matter/drug effects , Humans , Magnetic Resonance Imaging/trends , Male , Marijuana Abuse/epidemiology , Middle Aged , Neuroimaging/trends , Organ Size , Substance-Related Disorders/diagnostic imaging , Substance-Related Disorders/epidemiology , Nicotiana/adverse effects , Tobacco Use/epidemiology , Tobacco Use/trends , Tobacco Use Disorder/epidemiology , Young Adult
6.
J Mol Endocrinol ; 59(1): R11-R31, 2017 07.
Article in English | MEDLINE | ID: mdl-28400482

ABSTRACT

The hypothalamic-pituitary-adrenal (HPA) axis is critically involved in the neuroendocrine regulation of stress adaptation, and the restoration of homeostasis following stress exposure. Dysregulation of this axis is associated with stress-related pathologies like major depressive disorder, post-traumatic stress disorder, panic disorder and chronic anxiety. It has long been understood that stress during early life can have a significant lasting influence on the development of the neuroendocrine system and its neural regulators, partially by modifying epigenetic regulation of gene expression, with implications for health and well-being in later life. Evidence is accumulating that epigenetic plasticity also extends to adulthood, proposing it as a mechanism by which psychological trauma later in life can long-lastingly affect HPA axis function, brain plasticity, neuronal function and behavioural adaptation to neuropsychological stress. Further corroborating this claim is the phenomenon that these epigenetic changes correlate with the behavioural consequences of trauma exposure. Thereby, epigenetic modifications provide a putative molecular mechanism by which the behavioural phenotype and transcriptional/translational potential of genes involved in HPA axis regulation can change drastically in response to environmental challenges, and appear an important target for treatment of stress-related disorders. However, improved insight is required to increase their therapeutic (drug) potential. Here, we provide an overview of the growing body of literature describing the epigenetic modulation of the (primarily neuroendocrine) stress response as a consequence of adult life stress and interpret the implications for, and the challenges involved in applying this knowledge to, the identification and treatment of stress-related psychiatric disorders.


Subject(s)
Anxiety/metabolism , Depressive Disorder, Major/metabolism , Epigenesis, Genetic , Hypothalamo-Hypophyseal System/metabolism , Neurotransmitter Agents/metabolism , Pituitary-Adrenal System/metabolism , Animals , Anxiety/genetics , Anxiety/physiopathology , Brain/metabolism , Brain/physiopathology , DNA Methylation , Depressive Disorder, Major/genetics , Depressive Disorder, Major/physiopathology , Histones/genetics , Histones/metabolism , Homeostasis , Humans , Hypothalamo-Hypophyseal System/physiopathology , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/metabolism , Neurons/pathology , Pituitary-Adrenal System/physiopathology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
7.
Transl Psychiatry ; 6(5): e826, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27244235

ABSTRACT

Ibogaine is a naturally occurring substance which has been increasingly used in the lay-scene to reduce craving and relapse in patients with substance use disorders (SUDs). Although human clinical trials on the safety and efficacy of ibogaine are lacking, animal studies do support the efficacy of ibogaine. In this systematic review and meta-analysis (MA), we summarise these animal findings, addressing three questions: (1) does ibogaine reduce addictive behaviour in animal models of SUDs?; (2) what are the toxic effects of ibogaine on motor functioning, cerebellum and heart rhythm?; (3) what are neuropharmacological working mechanisms of ibogaine treatment in animal models of SUDs? MA of 27 studies showed that ibogaine reduced drug self-administration, particularly during the first 24 h after administration. Ibogaine had no effect on drug-induced conditioned place preference. Ibogaine administration resulted in motor impairment in the first 24 h after supplementation, and cerebral cell loss even weeks after administration. Data on ibogaines effect on cardiac rhythm, as well as on its neuropharmacological working mechanisms are limited. Our results warrant further studies into the clinical efficacy of ibogaine in SUD patients in reducing craving and substance use, but close monitoring of the patients is recommended because of the possible toxic effects. In addition, more work is needed to unravel the neuropharmacological working mechanisms of ibogaine and to investigate its effects on heart rhythm.


Subject(s)
Disease Models, Animal , Ibogaine/pharmacology , Illicit Drugs , Substance-Related Disorders/rehabilitation , Animals , Cerebellum/drug effects , Dose-Response Relationship, Drug , Female , Ibogaine/toxicity , Male , Motor Activity/drug effects , Neurons/drug effects , Self Administration
9.
Transl Psychiatry ; 5: e642, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26393488

ABSTRACT

The selective serotonin reuptake inhibitor (SSRI) fluoxetine is widely prescribed for the treatment of symptoms related to a variety of psychiatric disorders. After chronic SSRI treatment, some symptoms remediate on the long term, but the underlying mechanisms are not yet well understood. Here we studied the long-term consequences (40 days after treatment) of chronic fluoxetine exposure on genome-wide gene expression. During the treatment period, we measured body weight; and 1 week after treatment, cessation behavior in an SSRI-sensitive anxiety test was assessed. Gene expression was assessed in hippocampal tissue of adult rats using transcriptome analysis and several differentially expressed genes were validated in independent samples. Gene ontology analysis showed that upregulated genes induced by chronic fluoxetine exposure were significantly enriched for genes involved in myelination. We also investigated the expression of myelination-related genes in adult rats exposed to fluoxetine at early life and found two myelination-related genes (Transferrin (Tf) and Ciliary neurotrophic factor (Cntf)) that were downregulated by chronic fluoxetine exposure. Cntf, a neurotrophic factor involved in myelination, showed regulation in opposite direction in the adult versus neonatally fluoxetine-exposed groups. Expression of myelination-related genes correlated negatively with anxiety-like behavior in both adult and neonatally fluoxetine-exposed rats. In conclusion, our data reveal that chronic fluoxetine exposure causes on the long-term changes in expression of genes involved in myelination, a process that shapes brain connectivity and contributes to symptoms of psychiatric disorders.


Subject(s)
Behavior, Animal/drug effects , Ciliary Neurotrophic Factor/genetics , Fluoxetine/pharmacology , Hippocampus , Long Term Adverse Effects , Transferrin/genetics , Up-Regulation/drug effects , Animals , Gene Expression Profiling , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Long Term Adverse Effects/diagnosis , Long Term Adverse Effects/etiology , Long Term Adverse Effects/metabolism , Neural Conduction/drug effects , Neural Conduction/genetics , Pharmacogenetics , Rats , Selective Serotonin Reuptake Inhibitors/pharmacology
10.
Neuroscience ; 309: 243-58, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-25934041

ABSTRACT

Methylphenidate (MPH) is a widely prescribed stimulant drug for the treatment of attention deficit hyperactivity disorder (ADHD) in children and adolescents. Its use in this age group raises concerns regarding the potential interference with ongoing neurodevelopmental processes. Particularly the hippocampus is a highly plastic brain region that continues to develop postnatally and is involved in cognition and emotional behavior, functions known to be affected by MPH. In this study, we assessed whether hippocampal structure and function were affected by chronic oral MPH treatment and whether its effects were different in adolescent or adult rats. Using behavioral testing, resting-state functional MRI, post-mortem structural magnetic resonance imaging (MRI), and immunohistochemistry, we assessed MPH's effects on recognition memory, depressive-like behavior, topological features of functional connectivity networks, hippocampal shape and markers for hippocampal neurogenesis and proliferation. Object recognition memory was transiently impaired in adolescent treated rats, while in animals treated during adulthood, increased depressive-like behavior was observed. Neurogenesis was increased in adolescent treated rats, whereas cell proliferation was decreased following adult treatment. Adolescent treated rats showed inward shape deformations adjacent to ventral parahippocampal regions known to be involved in recognition memory, whereas such deformations were not observed in adult treated animals. Irrespective of the age of treatment, MPH affected topological features of ventral hippocampal functional networks. Thus, chronic oral treatment with a therapeutically relevant dose of MPH preferentially affected the ventral part of the hippocampus and induced contrasting effects in adolescent and adult rats. The differences in behavior were paralleled by opposite effects on adult neurogenesis and granule cell proliferation.


Subject(s)
Central Nervous System Stimulants/toxicity , Hippocampus/drug effects , Hippocampus/pathology , Methylphenidate/toxicity , Neurogenesis/drug effects , Administration, Oral , Aging/drug effects , Aging/pathology , Aging/physiology , Aging/psychology , Animals , Attention Deficit Disorder with Hyperactivity/drug therapy , Depressive Disorder/chemically induced , Depressive Disorder/pathology , Depressive Disorder/physiopathology , Hippocampus/growth & development , Hippocampus/physiology , Immunohistochemistry , Magnetic Resonance Imaging , Male , Neural Pathways/drug effects , Neural Pathways/growth & development , Neural Pathways/pathology , Neural Pathways/physiopathology , Neurogenesis/physiology , Rats, Wistar , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Rest
11.
Neurobiol Dis ; 80: 42-53, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25986729

ABSTRACT

Autism spectrum disorders are severe neurodevelopmental disorders, marked by impairments in reciprocal social interaction, delays in early language and communication, and the presence of restrictive, repetitive and stereotyped behaviors. Accumulating evidence suggests that dysfunction of the amygdala may be partially responsible for the impairment of social behavior that is a hallmark feature of ASD. Our studies suggest that a valproic acid (VPA) rat model of ASD exhibits an enlargement of the amygdala as compared to controls rats, similar to that observed in adolescent ASD individuals. Since recent research suggests that altered neuronal development and morphology, as seen in ASD, may result from a common post-transcriptional process that is under tight regulation by microRNAs (miRs), we examined genome-wide transcriptomics expression in the amygdala of rats prenatally exposed to VPA, and detected elevated miR-181c and miR-30d expression levels as well as dysregulated expression of their cognate mRNA targets encoding proteins involved in neuronal system development. Furthermore, selective suppression of miR-181c function attenuates neurite outgrowth and branching, and results in reduced synaptic density in primary amygdalar neurons in vitro. Collectively, these results implicate the small non-coding miR-181c in neuronal morphology, and provide a framework of understanding how dysregulation of a neurodevelopmentally relevant miR in the amygdala may contribute to the pathophysiology of ASD.


Subject(s)
Amygdala/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , MicroRNAs/metabolism , Amygdala/pathology , Animals , Autistic Disorder/chemically induced , Autistic Disorder/pathology , Disease Models, Animal , Neurons/metabolism , Neurons/pathology , Rats , Social Behavior , Transcriptome , Valproic Acid
12.
Transl Psychiatry ; 4: e409, 2014 Jul 08.
Article in English | MEDLINE | ID: mdl-25004389

ABSTRACT

The short allelic variant of the serotonin transporter (5-HTT) promoter-linked polymorphic region (5-HTTLPR) has been associated with the etiology of major depression by interaction with early life stress (ELS). Furthermore, 5-HTTLPR has been associated with abnormal functioning of the stress-responsive hypothalamo-pituitary-adrenal (HPA) axis. Here, we examined if, and at what level, the HPA-axis is affected in an animal model for ELS × 5-HTTLPR interactions. Heterozygous and homozygous 5-HTT knockout rats and their wild-type littermates were exposed daily at postnatal days 2-14 to 3 h of maternal separation. When grown to adulthood, plasma levels of adrenocorticotropic hormone (ACTH), and the major rat glucocorticoid, corticosterone (CORT), were measured. Furthermore, the gene expression of key HPA-axis players at the level of the hypothalamus, pituitary and adrenal glands was assessed. No 5-HTT genotype × ELS interaction effects on gene expression were observed at the level of the hypothalamus or pituitary. However, we found significant 5-HTT genotype × ELS interaction effects for plasma CORT levels and adrenal mRNA levels of the ACTH receptor, such that 5-HTT deficiency was associated under control conditions with increased, but after ELS with decreased basal HPA-axis activity. With the use of an in vitro adrenal assay, naïve 5-HTT knockout rats were furthermore shown to display increased adrenal ACTH sensitivity. Therefore, we conclude that basal HPA-axis activity is affected by the interaction of 5-HTT genotype and ELS, and is programmed, within the axis itself, predominantly at the level of the adrenal gland. This study therefore emphasizes the importance of the adrenal gland for HPA-related psychiatric disorders.


Subject(s)
Adrenal Glands/metabolism , Gene-Environment Interaction , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Stress, Psychological/metabolism , Adrenocorticotropic Hormone/blood , Age Factors , Animals , Corticosterone/blood , Disease Models, Animal , Maternal Deprivation , Rats , Rats, Transgenic , Receptors, Corticotropin/metabolism
14.
Prog Neuropsychopharmacol Biol Psychiatry ; 35(6): 1400-8, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-20883714

ABSTRACT

The selective serotonin reuptake inhibitor (SSRI) Prozac® (fluoxetine) is widely prescribed for the treatment of depression and anxiety-related disorders. While extensive research has established that fluoxetine is safe for adults, safety is not guaranteed for (unborn) children and adolescents. Some clinical studies have reported adverse outcomes, such as premature birth, neonatal cardiovascular abnormalities, and pulmonary hypertension in children whose mothers used SSRIs during pregnancy. In addition, several reports show that adolescent fluoxetine treatment increases risk for suicidal behavior. Despite these studies, fluoxetine is not contraindicated in the treatment of depressed pregnant women and adolescents. Longitudinal research in humans is limited because of ethical reasons and time constraints, and to overcome these limitations, rodents are used to increase insight in the age-dependent effects of fluoxetine exposure. It has been established that neonatal and adolescent fluoxetine exposure leads to paradoxical anxiety- and depression-like features in later life of rats and mice, although in some studies adolescent fluoxetine exposure was without effects. These age-dependent outcomes of fluoxetine may be explained by serotonin's neurotrophic effects, which may vary according to the developmental stage of the brain due to epigenetic modifications. Here we review the existing evidence for the age-dependent effects of fluoxetine in humans and rodents, address the gaps in our current knowledge and propose directions for future research. Given the overlap between human and rodent findings, rodents provide heuristic value in further research on the age-dependent effects of SSRIs.


Subject(s)
Anxiety/drug therapy , Depression/drug therapy , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Selective Serotonin Reuptake Inhibitors/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Age Factors , Animals , Anxiety/chemically induced , Behavior, Animal/drug effects , Behavior, Animal/physiology , Depression/chemically induced , Disease Models, Animal , Fluoxetine/adverse effects , Humans , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Serotonin/physiology , Selective Serotonin Reuptake Inhibitors/adverse effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
15.
Prog Neuropsychopharmacol Biol Psychiatry ; 35(6): 1357-75, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21129431

ABSTRACT

While modern neurobiology methods are necessary they are not sufficient to elucidate etiology and pathophysiology of affective disorders and develop new treatments. Achievement of these goals is contingent on applying cutting edge methods on appropriate disease models. In this review, the authors present four rodent models with good face-, construct-, and predictive-validity: the Flinders Sensitive rat line (FSL); the genetically "anxious" High Anxiety-like Behavior (HAB) line; the serotonin transporter knockout 5-HTT(-/-) rat and mouse lines; and the post-traumatic stress disorder (PTSD) model induced by exposure to predator scent, that they have employed to investigate the nature of depression and anxiety.


Subject(s)
Disease Models, Animal , Mental Disorders , Translational Research, Biomedical/methods , Animals , Anxiety , Depression , Humans , Mental Disorders/drug therapy , Mental Disorders/genetics , Mental Disorders/physiopathology , Mental Disorders/psychology
16.
Neurosci Biobehav Rev ; 34(3): 373-86, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19698744

ABSTRACT

The serotonin transporter knockout (SERT(-/-)) mouse, generated in 1998, was followed by the SERT(-/-) rat, developed in 2006. The availability of SERT(-/-) rodents creates the unique possibility to study the conservation of gene function across species. Here we summarize SERT(-/-) mouse and rat data, and discuss species (dis)similarities in neurobehavioral endophenotypes. Both SERT(-/-) rodent models show a disturbed serotonergic system, altered nociception, higher anxiety, decreased social behavior, as well as increased negative emotionality, behavioral inhibition and decision making. Used to model a wide range of psychiatric disorders, SERT(-/-) rodents may be particularly valuable in research on neurodevelopmental disorders such as depression, anxiety, and possibly autism. We conclude that SERT function is conserved across mice and rats and that their behavioral profile arises from common neurodevelopmental alterations. Because mice and rats have species-specific characteristics that confer differential research advantages, a comparison of the two models has heuristic value in understanding the mechanisms and behavioral outcome of SERT genetic variation in humans.


Subject(s)
Behavior, Animal/physiology , Brain/physiology , RNA-Binding Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Animals , Humans , Mice , RNA-Binding Proteins/genetics , Rats , Serotonin Plasma Membrane Transport Proteins/genetics
17.
Genes Brain Behav ; 8(8): 829-34, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19740092

ABSTRACT

Serotonin is well known for its role in affection, but less known for its role in cognition. The serotonin transporter (SERT) has an essential role in serotonergic neurotransmission as it determines the magnitude and duration of the serotonin signal in the synaptic cleft. There is evidence to suggest that homozygous SERT knockout rats (SERT(-/-)), as well as humans with the short SERT allele, show stronger cognitive effects than wild-type control rats (SERT(+/+)) and humans with the long SERT allele after acute tryptophan depletion. In rats, SERT genotype is known to affect brain serotonin levels, with SERT(-/-) rats having lower intracellular basal serotonin levels than wild-type rats in several brain areas. In the present study, it was investigated whether SERT genotype affects memory performance in an object recognition task with different inter-trial intervals. SERT(-/-), heterozygous SERT knockout (SERT(+/-)) and SERT(+/+) rats were tested in an object recognition test applying an inter-trial interval of 2, 4 and 8 h. SERT(-/-) and SERT(+/-) rats showed impaired object memory with an 8 h inter-trial interval, whereas SERT(+/+) rats showed intact object memory with this inter-trial interval. Although brain serotonin levels cannot fully explain the SERT genotype effect on object memory in rats, these results do indicate that serotonin is an important player in object memory in rats, and that lower intracellular serotonin levels lead to enhanced memory loss. Given its resemblance with the human SERT-linked polymorphic region and propensity to develop depression-like symptoms, our findings may contribute to further understanding of mechanisms underlying cognitive deficits in depression.


Subject(s)
Brain Chemistry/genetics , Genetic Predisposition to Disease/genetics , Memory Disorders/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin/metabolism , Animals , Brain/metabolism , Brain/physiopathology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Depressive Disorder/genetics , Depressive Disorder/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Gene Knockout Techniques , Genotype , Memory Disorders/metabolism , Memory Disorders/physiopathology , Rats , Recognition, Psychology/physiology , Serotonin Plasma Membrane Transport Proteins/metabolism
18.
Neuroscience ; 163(1): 308-15, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19527777

ABSTRACT

The nociceptin/orphanin FQ (N/OFQ) opioid peptide receptor (NOPr) is a new member of the opioid receptor family consisting of mu, delta and kappa opioid receptors. The anti-opioid properties of its endogenous ligand, N/OFQ provide the receptor interesting potentials in symptoms and processes related to drug addiction, learning and memory, anxiety and depression, and nociception. Using target-selected N-ethyl-N-nitrosourea (ENU)-driven mutagenesis we recently generated a rat model bearing a premature stop codon in the opioid-like receptor (oprl1) gene, and here we describe the primary characterization of this novel model. Data revealed that [(3)H]N/OFQ binding to brain slices was completely absent in rats homozygous for the premature stop codon (oprl1(-/-)). Heterozygous rats displayed an intermediate level of NOPr binding. Oprl1 receptor transcript levels, as determined by Northern blot analysis, were reduced by approximately 50% in oprl1(-/-) rats compared to wild-type controls (oprl1(+/+)), and no alternative spliced transcripts were observed. Quantitative autoradiographic mapping of mu, delta and kappa opioid receptors using [(3)H]DAMGO, [(3)H]deltorphin and [(3)H]CI-977, respectively, did not show any changes in opioid receptor binding. In conclusion, we present a novel mutant rat lacking NOPr without compensatory changes in mu, delta and kappa opioid receptors. We anticipate that this mutant rat will have heuristic value to further understand the function of NOPr.


Subject(s)
Binding, Competitive/genetics , Opioid Peptides/genetics , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Opioid/genetics , Alternative Splicing/genetics , Analgesics, Opioid/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain Chemistry/drug effects , Brain Chemistry/genetics , Codon, Nonsense/genetics , Disease Models, Animal , Gene Knockout Techniques , Organ Culture Techniques , Radioligand Assay , Rats , Rats, Wistar , Tritium , Nociceptin Receptor , Nociceptin
19.
Neuroscience ; 152(3): 573-84, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18295409

ABSTRACT

Human studies have shown that a reduction of 5-HT transporter (SERT) increases the vulnerability for anxiety and depression. Moreover, women are more vulnerable to develop depression and anxiety disorders than men. For that reason we hypothesized that homozygous 5-HT transporter knockout rat (SERT(-/-)) models, especially female, are valuable and reliable animal models for humans with an increased vulnerability for anxiety- and depression-related disorders. As rats are extensively used in neuroscience research, we used the unique 5-HT transporter knockout rat, that was recently generated using N-ethyl-N-nitrosurea (ENU) -driven mutagenesis, to test this hypothesis. Behavioral testing revealed that male and female SERT(-/-) rats spent less time in the center of the open field and spent less time on the open arm of the elevated plus maze compared with wild-type 5-HT transporter knockout rats (SERT(+/+)). In the novelty suppressed feeding test, only male SERT(-/-) rats showed a higher latency before starting to eat in a bright novel arena compared with SERT(+/+) controls. Both male and female SERT(-/-) rats showed a higher escape latency from their home cage than SERT(+/+) littermates. Moreover, SERT(-/-) rats were less mobile in the forced swim test, and sucrose consumption was reduced in SERT(-/-) rats relative to SERT(+/+) rats. Both effects were sex-independent. Neurochemically, basal extracellular 5-HT levels were elevated to a similar extent in male and female SERT(-/-) rats, which was not influenced by the selective 5-HT reuptake inhibitor citalopram. 5-HT immunostaining revealed no difference between SERT(+/+) and SERT(-/-) rats in the dorsal raphe nuclei, in both males and females. These findings demonstrate that SERT(-/-) rats show anxiety and depression-related behavior, independent of sex. Genetic inactivation of the SERT has apparently such a great impact on behavior, that hardly any differences are found between male and female rats. This knockout rat model may provide a valuable model to study anxiety- and depression-related disorders in male and female rats.


Subject(s)
Anxiety Disorders/genetics , Brain Chemistry/genetics , Depressive Disorder/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin/metabolism , Animals , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Appetite Regulation/genetics , Brain/metabolism , Brain/physiopathology , Depressive Disorder/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Down-Regulation/genetics , Exploratory Behavior/physiology , Extracellular Fluid/metabolism , Female , Male , Maze Learning/physiology , Microdialysis , Raphe Nuclei/metabolism , Rats , Rats, Mutant Strains , Reaction Time/genetics , Selective Serotonin Reuptake Inhibitors/pharmacology , Sex Characteristics , Synaptic Transmission/genetics
20.
Neuroscience ; 146(4): 1662-76, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17467186

ABSTRACT

Serotonergic signaling is involved in many neurobiological processes and disturbed 5-HT homeostasis is implicated in a variety of psychiatric and addictive disorders. Here, we describe the functional characterization of the serotonin transporter (SERT) knockout rat model, that is generated by N-ethyl-N-nitrosurea (ENU)-driven target-selected mutagenesis. Biochemical characterization revealed that SERT mRNA and functional protein are completely absent in homozygous knockout (SERT-/-) rats, and that there is a gene dose-dependent reduction in the expression and function of the SERT in heterozygous knockout rats. As a result, 5-HT homeostasis was found to be severely affected in SERT-/- rats: 5-HT tissue levels and depolarization-induced 5-HT release were significantly reduced, and basal extracellular 5-HT levels in the hippocampus were ninefold increased. Interestingly, we found no compensatory changes in in vitro activity of tryptophan hydroxylase and monoamine oxidase, the primary enzymes involved in 5-HT synthesis and degradation, respectively. Similarly, no major adaptations in non-serotonergic systems were found, as determined by dopamine and noradrenaline transporter binding, monoamine tissue levels, and depolarization-induced release of dopamine, noradrenaline, glutamate and GABA. In conclusion, neurochemical changes in the SERT knockout rat are primarily limited to the serotonergic system, making this novel rat model potentially very useful for studying the behavioral and neurobiological consequences of disturbed 5-HT homeostasis.


Subject(s)
Brain Chemistry/genetics , Serotonin Plasma Membrane Transport Proteins/deficiency , Serotonin/metabolism , Animals , Animals, Genetically Modified , Monoamine Oxidase/metabolism , Mutagenesis/drug effects , Mutagenesis/physiology , Neurotransmitter Agents/metabolism , Nitrosomethylurethane/pharmacology , Rats , Serotonin Plasma Membrane Transport Proteins/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Tryptophan Hydroxylase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...