Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Heliyon ; 8(1): e08692, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35024491

ABSTRACT

Optogenetics enables activation/silencing of specific neurons with unprecedented temporal and spatial resolution. The method, however, is prone to artefacts associated with biophysics of light used for illuminating opsin-expressing neurons. Here we employed Tph2-mhChR2-YFP transgenic mice, which express channelrhodopsin (ChR2) only in serotonergic neurons in the brain, to investigate behavioral effects of optogenetic stimulation of serotonergic neurons. Surprisingly, optogenetic stimulation enhanced locomotion even in ChR2-negative mice. Such unspecific effects are likely to be due to visual agitation caused by light leakage from the dental cement, which is commonly used to fixate optic fiber ferrules on the skull. When we employed black dental cement made by mixing carbons with dental cement powders, such unspecific effects were abolished in ChR2-negative mice, but not in ChR2-positive mice, confirming that enhanced locomotion resulted from serotonergic activation. The method allows extracting genuine behavioral effects of optogenetic stimulation without contamination from visual stimuli caused by light leakage.

2.
Front Neurosci ; 13: 245, 2019.
Article in English | MEDLINE | ID: mdl-31068767

ABSTRACT

Brain serotonin (5-hydroxytryptamine, 5-HT) system dysfunction is implicated in exaggerated fear responses triggering various anxiety-, stress-, and trauma-related disorders. However, the underlying mechanisms are not well understood. Here, we investigated the impact of constitutively inactivated 5-HT synthesis on context-dependent fear learning and extinction using tryptophan hydroxylase 2 (Tph2) knockout mice. Fear conditioning and context-dependent fear memory extinction paradigms were combined with c-Fos imaging and electrophysiological recordings in the dorsal hippocampus (dHip). Tph2 mutant mice, completely devoid of 5-HT synthesis in brain, displayed accelerated fear memory formation and increased locomotor responses to foot shock. Furthermore, recall of context-dependent fear memory was increased. The behavioral responses were associated with increased c-Fos expression in the dHip and resistance to foot shock-induced impairment of hippocampal long-term potentiation (LTP). In conclusion, increased context-dependent fear memory resulting from brain 5-HT deficiency involves dysfunction of the hippocampal circuitry controlling contextual representation of fear-related behavioral responses.

3.
Front Pharmacol ; 4: 97, 2013.
Article in English | MEDLINE | ID: mdl-23935583

ABSTRACT

Firing activity of serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN) is controlled by inhibitory somatodendritic 5-HT1A autoreceptors. This autoinhibitory mechanism is implicated in the etiology of disorders of emotion regulation, such as anxiety disorders and depression, as well as in the mechanism of antidepressant action. Here, we investigated how persistent alterations in brain 5-HT availability affect autoinhibition in two genetically modified mouse models lacking critical mediators of serotonergic transmission: 5-HT transporter knockout (Sert-/-) and tryptophan hydroxylase-2 knockout (Tph2-/-) mice. The degree of autoinhibition was assessed by loose-seal cell-attached recording in DRN slices. First, application of the 5-HT1A-selective agonist R(+)-8-hydroxy-2-(di-n-propylamino)tetralin showed mild sensitization and marked desensitization of 5-HT1A receptors in Tph2-/- mice and Sert-/- mice, respectively. While 5-HT neurons from Tph2-/- mice did not display autoinhibition in response to L-tryptophan, autoinhibition of these neurons was unaltered in Sert-/- mice despite marked desensitization of their 5-HT1A autoreceptors. When the Tph2-dependent 5-HT synthesis step was bypassed by application of 5-hydroxy-L-tryptophan (5-HTP), neurons from both Tph2-/- and Sert-/- mice decreased their firing rates at significantly lower concentrations of 5-HTP compared to wildtype controls. Our findings demonstrate that, as opposed to the prevalent view, sensitivity of somatodendritic 5-HT1A receptors does not predict the magnitude of 5-HT neuron autoinhibition. Changes in 5-HT1A receptor sensitivity may rather be seen as an adaptive mechanism to keep autoinhibition functioning in response to extremely altered levels of extracellular 5-HT resulting from targeted inactivation of mediators of serotonergic signaling.

4.
Curr Drug Targets ; 14(5): 549-70, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23547810

ABSTRACT

Disorders of emotion regulation such as anxiety disorders and depression are common and yet debilitating. Accumulating evidence suggests involvement of serotonin (5-HT) in the regulation of emotion. Mice with targeted deletion of genes encoding mediators of the serotonergic transmission have proven to be a powerful tool for understanding contributions of such mediators of emotion regulation. Over the last decade, research on mice with a targeted inactivation of the 5-HT transporter (5-Htt, Sert, Slc6a4) has considerably advanced our knowledge about functions that the 5-HTT plays in the context of emotion related to depression. Moreover, the recent advent of knockout (KO) mice for tryptophan hydroxylase 2 (Tph2 KO), which lacks the rate-limiting enzyme for 5-HT synthesis in the brain, has further provided insight to the brain serotonergic system and its role in emotion dysregulation. Here, we first highlight basic characteristics of the serotonergic system including the biosynthesis of 5-HT as well as the anatomy and firing activity of serotonergic neurons. Furthermore, characteristics of 5-Htt and Tph2 KO mice are covered together with association studies on human variants of 5-HTT and TPH2 in emotional regulation. Among various targets of serotonergic projections, which originate from the raphe nuclei in the brain stem, particular focus is placed on the hippocampus due to its unique dual role in memory and emotion. Finally, effects of therapeutic drugs and psychoactive drugs on KO mouse models as well as on synaptic plasticity will be discussed.


Subject(s)
Behavior, Animal , Depression/metabolism , Emotions , Hippocampus/metabolism , Serotonergic Neurons/metabolism , Serotonin Plasma Membrane Transport Proteins/deficiency , Serotonin/metabolism , Tryptophan Hydroxylase/deficiency , Animals , Cognition , Depression/genetics , Depression/physiopathology , Depression/psychology , Genetic Predisposition to Disease , Hippocampus/physiopathology , Humans , Mice , Mice, Knockout , Neuronal Plasticity , Phenotype , Serotonin Plasma Membrane Transport Proteins/genetics , Signal Transduction , Tryptophan Hydroxylase/genetics
5.
PLoS One ; 7(8): e43157, 2012.
Article in English | MEDLINE | ID: mdl-22912815

ABSTRACT

Brain serotonin (5-HT) is implicated in a wide range of functions from basic physiological mechanisms to complex behaviors, including neuropsychiatric conditions, as well as in developmental processes. Increasing evidence links 5-HT signaling alterations during development to emotional dysregulation and psychopathology in adult age. To further analyze the importance of brain 5-HT in somatic and brain development and function, and more specifically differentiation and specification of the serotonergic system itself, we generated a mouse model with brain-specific 5-HT deficiency resulting from a genetically driven constitutive inactivation of neuronal tryptophan hydroxylase-2 (Tph2). Tph2 inactivation (Tph2-/-) resulted in brain 5-HT deficiency leading to growth retardation and persistent leanness, whereas a sex- and age-dependent increase in body weight was observed in Tph2+/- mice. The conserved expression pattern of the 5-HT neuron-specific markers (except Tph2 and 5-HT) demonstrates that brain 5-HT synthesis is not a prerequisite for the proliferation, differentiation and survival of raphe neurons subjected to the developmental program of serotonergic specification. Furthermore, although these neurons are unable to synthesize 5-HT from the precursor tryptophan, they still display electrophysiological properties characteristic of 5-HT neurons. Moreover, 5-HT deficiency induces an up-regulation of 5-HT(1A) and 5-HT(1B) receptors across brain regions as well as a reduction of norepinephrine concentrations accompanied by a reduced number of noradrenergic neurons. Together, our results characterize developmental, neurochemical, neurobiological and electrophysiological consequences of brain-specific 5-HT deficiency, reveal a dual dose-dependent role of 5-HT in body weight regulation and show that differentiation of serotonergic neuron phenotype is independent from endogenous 5-HT synthesis.


Subject(s)
Brain/metabolism , Gene Silencing/physiology , Growth and Development/physiology , Raphe Nuclei/metabolism , Serotonin/deficiency , Tryptophan Hydroxylase/genetics , Age Factors , Animals , Autoradiography , Body Weight , Growth and Development/genetics , Histological Techniques , Hydroxyindoleacetic Acid/metabolism , Mice , Norepinephrine/metabolism , Receptors, Serotonin/metabolism , Sex Factors
6.
Philos Trans R Soc Lond B Biol Sci ; 367(1601): 2426-43, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22826343

ABSTRACT

Aggression, which comprises multi-faceted traits ranging from negative emotionality to antisocial behaviour, is influenced by an interaction of biological, psychological and social variables. Failure in social adjustment, aggressiveness and violence represent the most detrimental long-term outcome of neurodevelopmental disorders. With the exception of brain-specific tryptophan hydroxylase-2 (Tph2), which generates serotonin (5-HT) in raphe neurons, the contribution of gene variation to aggression-related behaviour in genetically modified mouse models has been previously appraised (Lesch 2005 Novartis Found Symp. 268, 111-140; Lesch & Merschdorf 2000 Behav. Sci. Law 18, 581-604). Genetic inactivation of Tph2 function in mice led to the identification of phenotypic changes, ranging from growth retardation and late-onset obesity, to enhanced conditioned fear response, increased aggression and depression-like behaviour. This spectrum of consequences, which are amplified by stress-related epigenetic interactions, are attributable to deficient brain 5-HT synthesis during development and adulthood. Human data relating altered TPH2 function to personality traits of negative emotionality and neurodevelopmental disorders characterized by deficits in cognitive control and emotion regulation are based on genetic association and are therefore not as robust as the experimental mouse results. Mouse models in conjunction with approaches focusing on TPH2 variants in humans provide unexpected views of 5-HT's role in brain development and in disorders related to negative emotionality, aggression and antisocial behaviour.


Subject(s)
Aggression/physiology , Antisocial Personality Disorder/physiopathology , Brain/physiopathology , Emotions/physiology , Serotonin/biosynthesis , Animals , Antisocial Personality Disorder/metabolism , Brain/metabolism , Cognition/physiology , Humans , Mice , Mice, Knockout , Phenotype , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin/genetics , Synaptic Transmission , Time Factors , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
7.
Psychoneuroendocrinology ; 36(3): 393-405, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21257271

ABSTRACT

Based on genetic variation, there is accumulating evidence that altered function of tryptophan hydroxylase-2 (TPH2), the enzyme critical for synthesis of serotonin (5-HT) in the brain, plays a role in anxiety-, aggression- and depression-related personality traits and in the pathogenesis of disorders featuring deficits in cognitive control and emotion regulation. Here, we appraise the genetic and neurobiological evidence to illustrate the critical role of TPH2 in central 5-HT system function and in the pathophysiology of a wide spectrum of disorders of cognitive control and emotion regulation, ranging from depression to attention-deficit/hyperactivity disorder (ADHD), a phenotype commonly associated with difficulties in the control of emotion and with a high co-morbidity of depression. Findings from psychophysiological and functional imaging studies are indicative of various TPH2 polymorphisms directly influencing serotonergic function and thus impacting on mood disorders and on the response to antidepressant treatment. Especially a combination with uncontrollable stress seems to potentiate these effects linking gene-environment interaction directly with behavioral dysfunction in human and animal models. TPH2-deficient mice display alterations in anxiety-like behavior which is accompanied by adaptational changes of 5-HT(1A) receptors and its associated signaling pathway. Mouse models in conjunction with cognitive neuroscience approaches in humans are providing unexpected results and it may well be that future research on TPH2 will provide an entirely new view of 5-HT in brain development and function related to neuropsychiatric disorders.


Subject(s)
Cognition Disorders/complications , Cognition Disorders/genetics , Mood Disorders/genetics , Tryptophan Hydroxylase/physiology , Animals , Disease Models, Animal , Emotions/physiology , Humans , Mice , Mice, Transgenic , Models, Biological , Mood Disorders/etiology , Tryptophan Hydroxylase/genetics
8.
BMC Evol Biol ; 9: 224, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19737383

ABSTRACT

BACKGROUND: The glutamate receptors (GluRs) play a vital role in the mediation of excitatory synaptic transmission in the central nervous system. To clarify the evolutionary dynamics and mechanisms of the GluR genes in the lineage leading to humans, we determined the complete sequences of the coding regions and splice sites of 26 chimpanzee GluR genes. RESULTS: We found that all of the reading frames and splice sites of these genes reported in humans were completely conserved in chimpanzees, suggesting that there were no gross structural changes in humans after their divergence from the human-chimpanzee common ancestor. We observed low KA/KS ratios in both humans and chimpanzees, and we found no evidence of accelerated evolution. We identified 30 human-specific "fixed" amino acid substitutions in the GluR genes by analyzing 80 human samples of seven different populations worldwide. Grantham's distance analysis showed that GRIN2C and GRIN3A are the most and the second most diverged GluR genes between humans and chimpanzees. However, most of the substitutions are non-radical and are not clustered in any particular region. Protein motif analysis assigned 11 out of these 30 substitutions to functional regions. Two out of these 11 substitutions, D71G in GRIN3A and R727H in GRIN3B, caused differences in the functional assignments of these genes between humans and other apes. CONCLUSION: We conclude that the GluR genes did not undergo drastic changes such as accelerated evolution in the human lineage after the divergence of chimpanzees. However, there remains a possibility that two human-specific "fixed" amino acid substitutions, D71G in GRIN3A and R727H in GRIN3B, are related to human-specific brain function.


Subject(s)
Amino Acid Substitution , Evolution, Molecular , Genome, Human , Pan troglodytes/genetics , Receptors, Glutamate/genetics , Animals , Comparative Genomic Hybridization , Humans , Multigene Family , Open Reading Frames , RNA Splice Sites , Selection, Genetic , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...