Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Insects ; 15(2)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38392513

ABSTRACT

The proboscis extension response (PER) has been widely used to evaluate honeybees' (Apis mellifera) learning and memory abilities, typically by using odors and visual cues for the conditioned stimuli. Here we asked whether honeybees could learn to distinguish between different magnitudes of the same type of stimulus, given as two speeds of air flux. By taking advantage of a novel automated system for administering PER experiments, we determined that the bees were highly successful when the lower air flux was rewarded and less successful when the higher flux was rewarded. Importantly, since our method includes AI-assisted analysis, we were able to consider subthreshold responses at a high temporal resolution; this analysis revealed patterns of rapid generalization and slowly acquired discrimination between the rewarded and unrewarded stimuli, as well as indications that the high air flux may have been mildly aversive. The learning curve for these mechanosensory stimuli, at least when the lower flux is rewarded, more closely mimics prior data from olfactory PER studies rather than visual ones, possibly in agreement with recent findings that the insect olfactory system is also sensitive to mechanosensory information. This work demonstrates a new modality to be used in PER experiments and lays the foundation for deeper exploration of honeybee cognitive processes when posed with complex learning challenges.

2.
Sci Rep ; 13(1): 3851, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36890201

ABSTRACT

Geosmin is an odorant produced by bacteria in moist soil. It has been found to be extraordinarily relevant to some insects, but the reasons for this are not yet fully understood. Here we report the first tests of the effect of geosmin on honey bees. A stinging assay showed that the defensive behaviour elicited by the bee's alarm pheromone component isoamyl acetate (IAA) is strongly suppressed by geosmin. Surprisingly, the suppression is, however, only present at very low geosmin concentrations, and disappears at higher concentrations. We investigated the underlying mechanisms at the level of the olfactory receptor neurons by means of electroantennography, finding the responses to mixtures of geosmin and IAA to be lower than to pure IAA, suggesting an interaction of both compounds at the olfactory receptor level. Calcium imaging of the antennal lobe (AL) revealed that neuronal responses to geosmin decreased with increasing concentration, correlating well with the observed behaviour. Computational modelling of odour transduction and coding in the AL suggests that a broader activation of olfactory receptor types by geosmin in combination with lateral inhibition could lead to the observed non-monotonic increasing-decreasing responses to geosmin and thus underlie the specificity of the behavioural response to low geosmin concentrations.


Subject(s)
Receptors, Odorant , Bees , Animals , Odorants , Pheromones/pharmacology , Naphthols
3.
Acta Neuropathol ; 144(1): 107-127, 2022 07.
Article in English | MEDLINE | ID: mdl-35551471

ABSTRACT

Mesial temporal lobe epilepsy with hippocampal sclerosis and a history of febrile seizures is associated with common variation at rs7587026, located in the promoter region of SCN1A. We sought to explore possible underlying mechanisms. SCN1A expression was analysed in hippocampal biopsy specimens of individuals with mesial temporal lobe epilepsy with hippocampal sclerosis who underwent surgical treatment, and hippocampal neuronal cell loss was quantitatively assessed using immunohistochemistry. In healthy individuals, hippocampal volume was measured using MRI. Analyses were performed stratified by rs7587026 type. To study the functional consequences of increased SCN1A expression, we generated, using transposon-mediated bacterial artificial chromosome transgenesis, a zebrafish line expressing exogenous scn1a, and performed EEG analysis on larval optic tecta at 4 day post-fertilization. Finally, we used an in vitro promoter analysis to study whether the genetic motif containing rs7587026 influences promoter activity. Hippocampal SCN1A expression differed by rs7587026 genotype (Kruskal-Wallis test P = 0.004). Individuals homozygous for the minor allele showed significantly increased expression compared to those homozygous for the major allele (Dunn's test P = 0.003), and to heterozygotes (Dunn's test P = 0.035). No statistically significant differences in hippocampal neuronal cell loss were observed between the three genotypes. Among 597 healthy participants, individuals homozygous for the minor allele at rs7587026 displayed significantly reduced mean hippocampal volume compared to major allele homozygotes (Cohen's D = - 0.28, P = 0.02), and to heterozygotes (Cohen's D = - 0.36, P = 0.009). Compared to wild type, scn1lab-overexpressing zebrafish larvae exhibited more frequent spontaneous seizures [one-way ANOVA F(4,54) = 6.95 (P < 0.001)]. The number of EEG discharges correlated with the level of scn1lab overexpression [one-way ANOVA F(4,15) = 10.75 (P < 0.001]. Finally, we showed that a 50 bp promoter motif containing rs7587026 exerts a strong regulatory role on SCN1A expression, though we could not directly link this to rs7587026 itself. Our results develop the mechanistic link between rs7587026 and mesial temporal lobe epilepsy with hippocampal sclerosis and a history of febrile seizures. Furthermore, we propose that quantitative precision may be important when increasing SCN1A expression in current strategies aiming to treat seizures in conditions involving SCN1A haploinsufficiency, such as Dravet syndrome.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Seizures, Febrile , Zebrafish Proteins/metabolism , Animals , Epilepsy/genetics , Epilepsy, Temporal Lobe/genetics , Genomics , Gliosis/pathology , Hippocampus/pathology , Humans , NAV1.1 Voltage-Gated Sodium Channel/genetics , Sclerosis/pathology , Seizures, Febrile/complications , Seizures, Febrile/genetics , Zebrafish
4.
Front Physiol ; 12: 790453, 2021.
Article in English | MEDLINE | ID: mdl-34950059

ABSTRACT

In insects, neuronal responses to clean air have so far been reported only episodically in moths. Here we present results obtained by fast two-photon calcium imaging in the honey bee Apis mellifera, indicating a substantial involvement of the antennal lobe, the first olfactory neuropil, in the processing of mechanical stimuli. Clean air pulses generate a complex pattern of glomerular activation that provides a code for stimulus intensity and dynamics with a similar level of stereotypy as observed for the olfactory code. Overlapping the air pulses with odor stimuli reveals a superposition of mechanosensory and odor response codes with high contrast. On the mechanosensitive signal, modulations were observed in the same frequency regime as the oscillatory motion of the antennae, suggesting a possible way to detect odorless airflow directions. The transduction of mechanosensory information via the insect antennae has so far been attributed primarily to Johnston's organ in the pedicel of the antenna. The possibility that the antennal lobe activation by clean air originates from Johnston's organ could be ruled out, as the signal is suppressed by covering the surfaces of the otherwise freely moving and bending antennae, which should leave Johnston's organ unaffected. The tuning curves of individual glomeruli indicate increased sensitivity at low-frequency mechanical oscillations as produced by the abdominal motion in waggle dance communication, suggesting a further potential function of this mechanosensory code. The discovery that the olfactory system can sense both odors and mechanical stimuli has recently been made also in mammals. The results presented here give hope that studies on insects can make a fundamental contribution to the cross-taxa understanding of this dual function, as only a few thousand neurons are involved in their brains, all of which are accessible by in vivo optical imaging.

5.
Neurosci Biobehav Rev ; 116: 1-20, 2020 09.
Article in English | MEDLINE | ID: mdl-32544542

ABSTRACT

Zebrafish are now widely accepted as a valuable animal model for a number of different central nervous system (CNS) diseases. They are suitable both for elucidating the origin of these disorders and the sequence of events culminating in their onset, and for use as a high-throughput in vivo drug screening platform. The availability of powerful and effective techniques for genome manipulation allows the rapid modelling of different genetic epilepsies and of conditions with seizures as a core symptom. With this review, we seek to summarize the current knowledge about existing epilepsy/seizures models in zebrafish (both pharmacological and genetic) and compare them with equivalent rodent and human studies. New findings obtained from the zebrafish models are highlighted. We believe that this comprehensive review will highlight the value of zebrafish as a model for investigating different aspects of epilepsy and will help researchers to use these models to their full extent.


Subject(s)
Epilepsy , Zebrafish , Animals , Disease Models, Animal , Epilepsy/genetics , Seizures
6.
Epilepsia ; 61(3): 549-560, 2020 03.
Article in English | MEDLINE | ID: mdl-32096222

ABSTRACT

OBJECTIVE: To pinpoint the earliest cellular defects underlying seizure onset (epileptogenic period) during perinatal brain development in a new zebrafish model of Dravet syndrome (DS) and to investigate potential disease-modifying activity of the 5HT2 receptor agonist fenfluramine. METHODS: We used CRISPR/Cas9 mutagenesis to introduce a missense mutation, designed to perturb ion transport function in all channel isoforms, into scn1lab, the zebrafish orthologue of SCN1A (encoding voltage-gated sodium channel alpha subunit 1). We performed behavioral analysis and electroencephalographic recordings to measure convulsions and epileptiform discharges, followed by single-cell RNA-Seq, morphometric analysis of transgenic reporter-labeled γ-aminobutyric acidergic (GABAergic) neurons, and pharmacological profiling of mutant larvae. RESULTS: Homozygous mutant (scn1labmut/mut ) larvae displayed spontaneous seizures with interictal, preictal, and ictal discharges (mean = 7.5 per 20-minute recording; P < .0001; one-way analysis of variance). Drop-Seq analysis revealed a 2:1 shift in the ratio of glutamatergic to GABAergic neurons in scn1labmut/mut larval brains versus wild type (WT), with dynamic changes in neuronal, glial, and progenitor cell populations. To explore disease pathophysiology further, we quantified dendritic arborization in GABAergic neurons and observed a 40% reduction in arbor number compared to WT (P < .001; n = 15 mutant, n = 16 WT). We postulate that the significant reduction in inhibitory arbors causes an inhibitory to excitatory neurotransmitter imbalance that contributes to seizures and enhanced electrical brain activity in scn1labmut/mut larvae (high-frequency range), with subsequent GABAergic neuronal loss and astrogliosis. Chronic fenfluramine administration completely restored dendritic arbor numbers to normal in scn1labmut/mut larvae, whereas similar treatment with the benzodiazepine diazepam attenuated seizures, but was ineffective in restoring neuronal cytoarchitecture. BrdU labeling revealed cell overproliferation in scn1labmut/mut larval brains that were rescued by fenfluramine but not diazepam. SIGNIFICANCE: Our findings provide novel insights into early mechanisms of DS pathogenesis, describe dynamic cell population changes in the scn1labmut/mut brain, and present first-time evidence for potential disease modification by fenfluramine.


Subject(s)
Brain/physiopathology , Epilepsies, Myoclonic/genetics , NAV1.1 Voltage-Gated Sodium Channel/genetics , Neuronal Plasticity/genetics , Zebrafish Proteins/genetics , Animals , Anticonvulsants/pharmacology , Brain/drug effects , Brain/metabolism , Brain/pathology , CRISPR-Cas Systems , Cell Proliferation/drug effects , Diazepam/pharmacology , Disease Models, Animal , Electroencephalography , Epilepsies, Myoclonic/metabolism , Epilepsies, Myoclonic/pathology , Epilepsies, Myoclonic/physiopathology , Fenfluramine/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gene Expression Profiling , Gliosis/genetics , Gliosis/pathology , Locomotion/drug effects , Mutation, Missense , NAV1.1 Voltage-Gated Sodium Channel/metabolism , Neuronal Plasticity/drug effects , RNA-Seq , Real-Time Polymerase Chain Reaction , Serotonin 5-HT2 Receptor Agonists/pharmacology , Single-Cell Analysis , Zebrafish , Zebrafish Proteins/metabolism
7.
Redox Biol ; 16: 237-247, 2018 06.
Article in English | MEDLINE | ID: mdl-29525604

ABSTRACT

DJ-1, a Parkinson's disease-associated protein, is strongly up-regulated in reactive astrocytes in Parkinson's disease. This is proposed to represent a neuronal protective response, although the mechanism has not yet been identified. We have generated a transgenic zebrafish line with increased astroglial DJ-1 expression driven by regulatory elements from the zebrafish GFAP gene. Larvae from this transgenic line are protected from oxidative stress-induced injuries as caused by MPP+, a mitochondrial complex I inhibitor shown to induce dopaminergic cells death. In a global label-free proteomics analysis of wild type and transgenic larvae exposed to MPP+, 3418 proteins were identified, in which 366 proteins were differentially regulated. In particular, we identified enzymes belonging to primary metabolism to be among proteins affected by MPP+ in wild type animals, but not affected in the transgenic line. Moreover, by performing protein profiling on isolated astrocytes we showed that an increase in astrocytic DJ-1 expression up-regulated a large group of proteins associated with redox regulation, inflammation and mitochondrial respiration. The majority of these proteins have also been shown to be regulated by Nrf2. These findings provide a mechanistic insight into the protective role of astroglial up-regulation of DJ-1 and show that our transgenic zebrafish line with astrocytic DJ-1 over-expression can serve as a useful animal model to understand astrocyte-regulated neuroprotection associated with oxidative stress-related neurodegenerative disease.


Subject(s)
Inflammation/genetics , NF-E2-Related Factor 2/genetics , Parkinson Disease/genetics , Protein Deglycase DJ-1/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified/genetics , Astrocytes/metabolism , Astrocytes/pathology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Gene Expression Regulation, Enzymologic , Humans , Inflammation/pathology , Larva/genetics , Mitochondria/genetics , Mitochondria/pathology , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/metabolism , Oxidation-Reduction , Oxidative Stress , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Deglycase DJ-1/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism
8.
J Hum Genet ; 60(7): 399-401, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25877001

ABSTRACT

Genetic studies of complex traits have become increasingly successful as progress is made in next-generation sequencing. We aimed at discovering single nucleotide variation present in known and new candidate genes for developmental dyslexia: CYP19A1, DCDC2, DIP2A, DYX1C1, GCFC2 (also known as C2orf3), KIAA0319, MRPL19, PCNT, PRMT2, ROBO1 and S100B. We used next-generation sequencing to identify single-nucleotide polymorphisms in the exons of these 11 genes in pools of 100 DNA samples of Finnish individuals with developmental dyslexia. Subsequent individual genotyping of those 100 individuals, and additional cases and controls from the Finnish and German populations, validated 92 out of 111 different single-nucleotide variants. A nonsynonymous polymorphism in DCDC2 (corrected P = 0.002) and a noncoding variant in S100B (corrected P = 0.016) showed a significant association with spelling performance in families of German origin. No significant association was found for the variants neither in the Finnish case-control sample set nor in the Finnish family sample set. Our findings further strengthen the role of DCDC2 and implicate S100B, in the biology of reading and spelling.


Subject(s)
Dyslexia/genetics , Microtubule-Associated Proteins/genetics , S100 Calcium Binding Protein beta Subunit/genetics , Case-Control Studies , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
9.
Neural Plast ; 2013: 605079, 2013.
Article in English | MEDLINE | ID: mdl-23936678

ABSTRACT

The nervous system is highly sensitive to experience during early postnatal life, but this phase of heightened plasticity decreases with age. Recent studies have demonstrated that developmental-like plasticity can be reactivated in the visual cortex of adult animals through environmental or pharmacological manipulations. These findings provide a unique opportunity to study the cellular and molecular mechanisms of adult plasticity. Here we used the monocular deprivation paradigm to investigate large-scale gene expression patterns underlying the reinstatement of plasticity produced by fluoxetine in the adult rat visual cortex. We found changes, confirmed with RT-PCRs, in gene expression in different biological themes, such as chromatin structure remodelling, transcription factors, molecules involved in synaptic plasticity, extracellular matrix, and excitatory and inhibitory neurotransmission. Our findings reveal a key role for several molecules such as the metalloproteases Mmp2 and Mmp9 or the glycoprotein Reelin and open up new insights into the mechanisms underlying the reopening of the critical periods in the adult brain.


Subject(s)
Gene Expression/physiology , Neuronal Plasticity/physiology , Sensory Deprivation/physiology , Visual Cortex/physiology , Animals , Fluoxetine/pharmacology , Gene Expression/drug effects , Neuronal Plasticity/drug effects , Rats , Rats, Long-Evans , Reelin Protein , Selective Serotonin Reuptake Inhibitors/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Visual Cortex/drug effects
10.
J Physiol ; 590(19): 4777-87, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22674715

ABSTRACT

There is evidence that developmental-like plasticity can be reactivated in the adult visual cortex. Although activity-dependent transcription factors underlying the process of plasticity reactivation are currently unknown, recent studies point towards NPAS4 as a candidate gene for the occurrence of plasticity in the adult visual system. Here, we addressed whether NPAS4 is involved in the reinstatement of plasticity by using the monocular deprivation protocol and long-term fluoxetine treatment as a pharmacological strategy that restores plasticity in adulthood. A combination of molecular assays for gene expression and epigenetic analysis, gene delivery by lentiviral infection, shRNA interference and electrophysiology as a functional read-out, revealed a previously unknown role for the transcription factor NPAS4 in the regulation of adult visual cortical plasticity. We found that NPAS4 overexpression restores ocular dominance plasticity in adult naive animals whereas NPAS4 down-regulation prevents the plastic outcome caused by fluoxetine in adulthood.Our findings lead the way to the identification of novel therapeutic targets for pathological conditions where reorganization of neuronal networks would be beneficial in adult life.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Neuronal Plasticity/physiology , Visual Cortex/physiology , Animals , DNA Methylation , Evoked Potentials, Visual/drug effects , Fluoxetine/pharmacology , Neuronal Plasticity/drug effects , Promoter Regions, Genetic , Rats , Rats, Long-Evans , Selective Serotonin Reuptake Inhibitors/pharmacology , Visual Cortex/drug effects
11.
Neural Plast ; 2012: 250421, 2012.
Article in English | MEDLINE | ID: mdl-22720172

ABSTRACT

The central nervous system architecture is markedly modified by sensory experience during early life, but a decline of plasticity occurs with age. Recent studies have challenged this dogma providing evidence that both pharmacological treatments and paradigms based on the manipulation of environmental stimulation levels can be successfully employed as strategies for enhancing plasticity in the adult nervous system. Insulin-like growth factor 1 (IGF-1) is a peptide implicated in prenatal and postnatal phases of brain development such as neurogenesis, neuronal differentiation, synaptogenesis, and experience-dependent plasticity. Here, using the visual system as a paradigmatic model, we report that IGF-1 reactivates neural plasticity in the adult brain. Exogenous administration of IGF-1 in the adult visual cortex, indeed, restores the susceptibility of cortical neurons to monocular deprivation and promotes the recovery of normal visual functions in adult amblyopic animals. These effects were accompanied by a marked reduction of intracortical GABA levels. Moreover, we show that a transitory increase of IGF-1 expression is associated to the plasticity reinstatement induced by environmental enrichment (EE) and that blocking IGF-1 action by means of the IGF-1 receptor antagonist JB1 prevents EE effects on plasticity processes.


Subject(s)
Aging/physiology , Insulin-Like Growth Factor I/physiology , Neuronal Plasticity/physiology , Sensory Deprivation/physiology , Visual Cortex/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Microdialysis/methods , Nerve Net/metabolism , Nerve Net/physiology , Neural Inhibition/physiology , Rats , Rats, Long-Evans , Visual Cortex/drug effects
12.
Eur J Pharmacol ; 687(1-3): 9-13, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22546227

ABSTRACT

Earlier findings suggest that, in addition to its well-known neurotrophic role, brain-derived neurotrophic factor (BDNF) is also involved in the rewarding and reinforcing effects of drugs of abuse. The purpose of the present study was to examine the effects of acute administration of ethanol (1.25 or 2.5 g/kg i.p.) on the expression profile of BDNF in the rat brain by determining the BDNF mRNA expression in the frontal cortex, nucleus accumbens, amygdala, hippocampus, and ventral tegmental area. Ethanol decreased BDNF mRNA levels dose-dependently in the hippocampus, and after the higher ethanol dose in the frontal cortex, nucleus accumbens and amygdala, while increasing them in the ventral tegmental area. Furthermore, BDNF mRNA expression was found to be regulated in a temporally different manner in all investigated brain areas. These data suggest that BDNF is involved in the acute effects of ethanol, but separate brain areas may be differentially engaged in the mediation of these effects.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Ethanol/administration & dosage , Gene Expression Regulation/drug effects , Animals , Ethanol/blood , Ethanol/pharmacokinetics , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar
13.
Science ; 334(6063): 1731-4, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22194582

ABSTRACT

Antidepressant drugs and psychotherapy combined are more effective in treating mood disorders than either treatment alone, but the neurobiological basis of this interaction is unknown. To investigate how antidepressants influence the response of mood-related systems to behavioral experience, we used a fear-conditioning and extinction paradigm in mice. Combining extinction training with chronic fluoxetine, but neither treatment alone, induced an enduring loss of conditioned fear memory in adult animals. Fluoxetine treatment increased synaptic plasticity, converted the fear memory circuitry to a more immature state, and acted through local brain-derived neurotrophic factor. Fluoxetine-induced plasticity may allow fear erasure by extinction-guided remodeling of the memory circuitry. Thus, the pharmacological effects of antidepressants need to be combined with psychological rehabilitation to reorganize networks rendered more plastic by the drug treatment.


Subject(s)
Antidepressive Agents, Second-Generation/therapeutic use , Anxiety Disorders/therapy , Behavior Therapy , Extinction, Psychological , Fear , Fluoxetine/therapeutic use , Neuronal Plasticity/drug effects , Amygdala/cytology , Amygdala/drug effects , Amygdala/physiology , Animals , Antidepressive Agents, Second-Generation/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Combined Modality Therapy , Conditioning, Classical , Excitatory Postsynaptic Potentials/drug effects , Fluoxetine/pharmacology , Interneurons/drug effects , Interneurons/physiology , Male , Memory , Mice , Mice, Inbred C57BL , Nerve Net/drug effects , Nerve Net/physiology , Neurons/cytology , Neurons/drug effects , Synaptic Transmission/drug effects
14.
Eur J Neurosci ; 33(1): 49-57, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21156002

ABSTRACT

Cortical circuitries are highly sensitive to experience during early life but this phase of heightened plasticity decreases with development. We recently demonstrated that fluoxetine reinstates a juvenile-like form of plasticity in the adult visual system. Here we explored cellular and molecular mechanisms that underlie the occurrence of these plastic phenomena. Adult rats were intracortically treated with serotonin (5-HT) whereas long-term fluoxetine-treated rats were infused with the 5-HT(1A) -receptor antagonist WAY-100635, brain-derived neurotrophic factor (BDNF) scavenger trkB-IgG or the mitogen-activated protein kinase inhibitor U0126. Plasticity was assessed as variations of visual cortex responsiveness after unilateral eyelid suture and reverse occlusion by using an electrophysiological approach. Real-time PCR and chromatin immunoprecipitation analysis were then used to explore alterations in gene expression and modifications of chromatin structure associated with the plastic outcome caused by fluoxetine in the visual system. Local infusion of 5-HT into visual cortex restored susceptibility to monocular deprivation in adulthood whereas infusion of WAY-100635, trkB-IgG or U0126 prevented the process of plasticity reactivation in fluoxetine-treated animals. Long-term fluoxetine treatment promoted a transient increase of Bdnf expression in the visual cortex, which was paralleled by an increased histone acetylation status at Bdnf promoter regions and by decreased expression of Hdac5. Accordingly, enhancing histone acetylation levels by systemic treatment with Trichostatin-A reactivated plasticity in the adult while WAY-100635-infusion prevented epigenetic modifications in Bdnf promoter areas. The data suggest a key role for 5-HT(1A) receptor and BDNF-trkB signalling in driving a transitory epigenetic remodelling of chromatin structure that underlies the reactivation of plasticity in the visual system.


Subject(s)
Epigenesis, Genetic/drug effects , Neuronal Plasticity/drug effects , Serotonin/pharmacology , Visual Cortex/drug effects , Visual Cortex/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Butadienes/pharmacology , Chromatin/metabolism , Chromatin/ultrastructure , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/physiology , Fluoxetine/pharmacology , Neuronal Plasticity/physiology , Nitriles/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Long-Evans , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, trkB/metabolism , Sensory Deprivation , Serotonin Antagonists/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Signal Transduction/physiology
15.
Int J Neuropsychopharmacol ; 12(10): 1367-81, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19400982

ABSTRACT

Converging evidence points to adaptive changes in neuroplasticity and gene expression as mediators of therapeutic action of antidepressants. Activation of cAMP response-element binding protein (CREB) and CREB-regulating signalling are considered main effectors in these mechanisms. We analysed the temporal profile of intracellular changes induced by antidepressants, by measuring activation of major CREB-regulating signalling cascades and activation (Ser133 phosphorylation) of CREB. The main aims of the study were to investigate how these different variables are modulated with time, whether stronger activation of signalling cascades corresponds to stronger activation of CREB, and whether these changes are different in distinct brain areas. Rat groups were treated for 1, 2 or 3 wk with the antidepressants fluoxetine or reboxetine; in additional groups drug treatment was followed by a washout week (3+1). Activation of CREB and major effectors in signalling cascades were analysed by Western blot analysis with phospho-antibodies, in nuclear and cytosolic fractions from hippocampus and prefrontal/frontal cortex (P/FC). Surprisingly, CREB activation was already maximal after 1-wk treatment. In hippocampus early and stronger CREB activation was consistent with early and stronger activation of signalling. For both drugs, the profile of activation in P/FC was different from that observed in hippocampus. The results also showed that, contrary to the activatory role of MAP-ERKs and CaM kinase IV, nuclear alphaCaM kinase II was inactivated in parallel with activation of CREB.


Subject(s)
Antidepressive Agents/pharmacology , Cyclic AMP Response Element-Binding Protein/biosynthesis , Signal Transduction/physiology , Animals , Cyclic AMP Response Element-Binding Protein/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Time Factors
16.
BMC Neurosci ; 9: 61, 2008 Jul 05.
Article in English | MEDLINE | ID: mdl-18601743

ABSTRACT

BACKGROUND: Recent rodent studies reported that antidepressant treatments affect the expression of brain-derived neurotrophic factor (BDNF) mRNA in a way that is dependent on treatment duration, by selective modulation of different BDNF transcripts. However, no data are available for the human BDNF gene. We studied the effect of different antidepressants on BDNF mRNA expression in human neuroblastoma SH-SY5Y cells. RESULTS: Cultured cells were treated with the antidepressants fluoxetine, reboxetine and desipramine for different time lengths (6, 24, 48 hours). Expression of total BDNF mRNA was analyzed by reverse transcription PCR and levels of different BDNF transcripts were detected by hemi-nested PCR with specific primers. Short-term treatment (6 hours) with reboxetine or desipramine reduced total BDNF, whereas long-term treatment (48 hours) significantly increased total BDNF mRNA levels. These changes were accounted for by differential regulation of BDNF IV and VIa/b transcripts. Fluoxetine showed no significant effects. CONCLUSION: This is the first study showing biphasic changes in the expression of total and specific BDNF transcripts in human cells following antidepressant treatments. These findings suggest that biphasic induction of BDNF by antidepressants could be a feature common to rodents and humans and encourage the use of SH-SY5Y cells as a tool for investigation of drug effects on human genes.


Subject(s)
Antidepressive Agents/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Gene Expression Regulation, Neoplastic/drug effects , Antidepressive Agents, Second-Generation/pharmacology , Cell Line, Tumor , Desipramine/pharmacology , Fluoxetine/pharmacology , Humans , Morpholines/pharmacology , Neuroblastoma/genetics , Neuroblastoma/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reboxetine , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
17.
Neuropsychopharmacology ; 32(12): 2511-9, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17356571

ABSTRACT

Changes in synaptic plasticity are involved in pathophysiology of depression and in the mechanism of antidepressants. Ca(2+)/calmodulin (CaM) kinase II, a protein kinase involved in synaptic plasticity, has been previously shown to be a target of antidepressants. We previously found that antidepressants activate the kinase in hippocampal neuronal cell bodies by increasing phosphorylation at Thr(286), reduce the kinase phosphorylation in synaptic membranes, and in turn its phosphorylation-dependent interaction with syntaxin-1 and the release of glutamate from hippocampal synaptosomes. Here, we investigated the chronic effect of different antidepressants (fluoxetine, desipramine, and reboxetine) on the expression and function of the kinase in distinct subcellular compartments in order to dissect the different kinase pools affected. Acute treatments did not induce any change in the kinase. In total tissue extracts chronic drug treatments induced activation of the kinase; in hippocampus (HC), but not in prefrontal/frontal cortex, this was partially accounted for by increased Thr(286) phosphorylation, suggesting the involvement of different mechanisms of activation. In synaptosomes, all drugs reduced the kinase phosphorylation, particularly in HC where, upon fractionation of the synaptosomal particulate into synaptic vesicles and membranes, we found that the drugs induced a redistribution and differential activation of the kinase between membranes and vesicles. Furthermore, a large decrease in the level and phosphorylation of synapsin I located at synaptic membranes was consistent with the observed decrease of CaM kinase II. Overall, antidepressants induce a complex pattern of modifications in distinct subcellular compartments; at presynaptic level, these changes are in line with a dampening of glutamate release.


Subject(s)
Antidepressive Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Synaptosomes/drug effects , Synaptosomes/enzymology , Animals , Cadherins/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Hippocampus/ultrastructure , Male , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Subcellular Fractions/drug effects , Synaptophysin/metabolism
18.
Int J Neuropsychopharmacol ; 10(4): 491-6, 2007 Aug.
Article in English | MEDLINE | ID: mdl-16923323

ABSTRACT

Lithium is widely used in the treatment of bipolar disorder, although its mechanism of action is not fully clear. This study was undertaken to assess the effects of prolonged lithium administration on cAMP responsive element-binding protein (CREB) phosphorylation and CaM kinase IV (CaMKIV), one of the main kinases phosphorylating CREB in neurons following synaptic activation. CREB total protein expression and phosphorylation (Ser133), as well as CaMKIV enzymatic activity, phosphorylation of Thr196 (the activator residue) and kinase expression level were assessed in total homogenates and nuclei from the hippocampus and prefrontal/frontal cortex following 5 wk lithium treatment. Whereas no significant effects were found in prefrontal/frontal cortex, lithium administration reduced CREB phosphorylation and at the same time down-regulated CaMKIV (enzymatic activity, phospho-Thr196 and protein expression level) in cell nuclei from the hippocampus. These data suggest for the first time the involvement of CaMKIV in the mechanism of action of lithium.


Subject(s)
Antimanic Agents/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Hippocampus/drug effects , Lithium Carbonate/pharmacology , Prefrontal Cortex/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4 , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Down-Regulation , Hippocampus/enzymology , Hippocampus/metabolism , Male , Phosphorylation , Prefrontal Cortex/enzymology , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
19.
Pharmacol Rev ; 58(1): 115-34, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16507885

ABSTRACT

Regulation of gene expression represents a major component in antidepressant drug action. The effect of antidepressant treatments on the function of cAMP-responsive element binding protein (CREB), a transcription factor that regulates expression of several genes involved in neuroplasticity, cell survival, and cognition, has been extensively studied. Although there is general agreement that chronic antidepressants stimulate CREB function, conflicting results suggest that different effects may depend on drug type, drug dosage, and different experimental paradigms. CREB function is activated by a vast array of physiological stimuli, conveyed through a number of signaling pathways acting in concert, but thus far the effects of antidepressants on CREB have been analyzed mostly with regard to the cAMP-protein kinase A pathway. A growing body of data shows that other major pathways, such as the calcium/calmodulin-dependent kinase and the mitogen-activated kinase cascades, are involved in activity-dependent regulation of gene expression and may also be implicated in the mechanism of action of antidepressants. In this article the available evidence is reviewed with an attempt to identify the reasons for experimental discrepancies and possible directions for future research. Particularemphasis is given to the regulation of brain-derived neurotrophic factor (BDNF), a CREB-regulated gene, which has been implicated in both the pathophysiology and pharmacology of mood disorders. The array of different results obtained by various groups is analyzed with an eye on recent advancements in the regulation of BDNF transcription, in an attempt to understand better the mechanisms of drug action and dissect molecular requirements for faster and more efficient antidepressant treatment.


Subject(s)
Antidepressive Agents/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Gene Expression Regulation/drug effects , Humans , Neuronal Plasticity , Protein Kinases/metabolism , Signal Transduction
20.
Biol Psychiatry ; 59(8): 713-20, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16460696

ABSTRACT

BACKGROUND: Several reports have shown that the glutamatergic system is involved in both the pathogenesis of affective and stress-related disorders and in the action of antidepressant drugs. In particular, antidepressant treatment was shown to modulate expression and function of ionotropic glutamate receptors, to inhibit glutamate release and to restore synaptic plasticity impaired by stress. METHODS: We analyzed the mRNA expression and RNA editing of alpha-amino-propionic-acid (AMPA) and kainate (KA) receptor subunits, in the pre-frontal/frontal cortex (P/FC) and hippocampus (HI) of rats chronically treated with three different drugs: the selective serotonin (5-HT) reuptake inhibitor fluoxetine, the selective noradrenaline (NA) reuptake inhibitor reboxetine and the tricyclic antidepressant desipramine. RESULTS: Our data showed that fluoxetine and desipramine exerted moderate but selective effects on glutamate receptor expression and editing, while reboxetine appeared to be the drug that affects glutamate receptors (GluR) most. The most consistent effect, observed with pronoradrenergic drugs (desipramine and reboxetine), was a decrease of GluR3 expression both in P/FC and HI. Interestingly, in HI, the same drugs also decreased the editing levels of either the flip (desipramine) or flop (reboxetine) form of GluR3. CONCLUSIONS: Overall, these results point to specific and regionally discrete changes in the expression and editing level of glutamate receptors and, in particular, to a selective reduction of conductance for GluR3-containing receptors following treatment with antidepressant drugs. These data support the hypothesis that changes in glutamate neurotransmission are involved in the therapeutic effects induced by these drugs.


Subject(s)
Antidepressive Agents/pharmacology , Gene Expression/drug effects , RNA Editing/drug effects , Receptors, AMPA/metabolism , Receptors, Kainic Acid/metabolism , Animals , Brain/anatomy & histology , Brain/drug effects , Brain/metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, AMPA/classification , Receptors, AMPA/genetics , Receptors, Kainic Acid/classification , Receptors, Kainic Acid/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...