Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
Más filtros










Intervalo de año de publicación
1.
Cereb Cortex ; 34(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39128940

RESUMEN

The orbitofrontal cortex and amygdala collaborate in outcome-guided decision-making through reciprocal projections. While serotonin transporter knockout (SERT-/-) rodents show changes in outcome-guided decision-making, and in orbitofrontal cortex and amygdala neuronal activity, it remains unclear whether SERT genotype modulates orbitofrontal cortex-amygdala synchronization. We trained SERT-/- and SERT+/+ male rats to execute a task requiring to discriminate between two auditory stimuli, one predictive of a reward (CS+) and the other not (CS-), by responding through nose pokes in opposite-side ports. Overall, task acquisition was not influenced by genotype. Next, we simultaneously recorded local field potentials in the orbitofrontal cortex and amygdala of both hemispheres while the rats performed the task. Behaviorally, SERT-/- rats showed a nonsignificant trend for more accurate responses to the CS-. Electrophysiologically, orbitofrontal cortex-amygdala synchronization in the beta and gamma frequency bands during response selection was significantly reduced and associated with decreased hubness and clustering coefficient in both regions in SERT-/- rats compared to SERT+/+ rats. Conversely, theta synchronization at the time of behavioral response in the port associated with reward was similar in both genotypes. Together, our findings reveal the modulation by SERT genotype of the orbitofrontal cortex-amygdala functional connectivity during an auditory discrimination task.


Asunto(s)
Amígdala del Cerebelo , Discriminación en Psicología , Ritmo Gamma , Corteza Prefrontal , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Animales , Masculino , Corteza Prefrontal/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Amígdala del Cerebelo/fisiología , Ritmo Gamma/fisiología , Ratas , Discriminación en Psicología/fisiología , Ritmo beta/fisiología , Vías Nerviosas/fisiología , Recompensa , Percepción Auditiva/fisiología , Estimulación Acústica , Ratas Transgénicas
2.
Int J Mol Sci ; 25(8)2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38674044

RESUMEN

Serotonin transporter (SERT) deficiency has been implicated in metabolic syndrome, intestinal inflammation, and microbial dysbiosis. Interestingly, changes in microbiome metabolic capacity and several alterations in host gene expression, including lipid metabolism, were previously observed in SERT-/- mice ileal mucosa. However, the precise host or microbial metabolites altered by SERT deficiency that may contribute to the pleiotropic phenotype of SERT KO mice are not yet understood. This study investigated the hypothesis that SERT deficiency impacts lipid and microbial metabolite abundances in the ileal mucosa, where SERT is highly expressed. Ileal mucosal metabolomics was performed by Metabolon on wild-type (WT) and homozygous SERT knockout (KO) mice. Fluorescent-activated cell sorting (FACS) was utilized to measure immune cell populations in ileal lamina propria to assess immunomodulatory effects caused by SERT deficiency. SERT KO mice exhibited a unique ileal mucosal metabolomic signature, with the most differentially altered metabolites being lipids. Such changes included increased diacylglycerols and decreased monoacylglycerols in the ileal mucosa of SERT KO mice compared to WT mice. Further, the ileal mucosa of SERT KO mice exhibited several changes in microbial-related metabolites known to play roles in intestinal inflammation and insulin resistance. SERT KO mice also had a significant reduction in the abundance of ileal group 3 innate lymphoid cells (ILC3). In conclusion, SERT deficiency induces complex alterations in the ileal mucosal environment, indicating potential links between serotonergic signaling, gut microbiota, mucosal immunity, intestinal inflammation, and metabolic syndrome.


Asunto(s)
Microbioma Gastrointestinal , Íleon , Mucosa Intestinal , Ratones Noqueados , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Animales , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Íleon/metabolismo , Íleon/patología , Mucosa Intestinal/metabolismo , Ratones , Metabolismo de los Lípidos , Metabolómica/métodos , Masculino , Metaboloma , Ratones Endogámicos C57BL
3.
Placenta ; 115: 158-168, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34649169

RESUMEN

INTRODUCTION: The mouse placenta accumulates and possibly produces serotonin (5-hydroxytryptamine; 5-HT) in parietal trophoblast giant cells (pTGC) located at the interface between the placenta and maternal deciduum. However, the roles of 5-HT in placental function are unclear. This lack of information is unfortunate, given that selective serotonin-reuptake inhibitors are commonly used to combat depression in pregnant women. The high affinity 5-HT transporter SLC6A4 (also known as SERT) is the target of such drugs and likely controls much of 5-HT uptake into pTGC and other placental cells. We hypothesized that ablation of the Slc6a4 gene would result in morphological changes correlated with placental gene expression changes, especially for those involved in nutrient acquisition and metabolism, and thereby, provide insights into 5-HT placental function. METHODS: Placentas were collected at embryonic age (E) 12.5 from Slc6a4 knockout (KO) and wild-type (WT) conceptuses. Histological analyses, RNAseq, qPCR, and integrative correlation analyses were performed. RESULTS: Slc6a4 KO placentas had a considerable increased pTGC to spongiotrophoblast area ratio relative to WT placentas and significantly elevated expression of genes associated with intestinal functions, including nutrient sensing, uptake, and catabolism, and blood clotting. Integrative correlation analyses revealed upregulation of many of these genes was correlated with pTGC layer expansion. One other key gene was dopa decarboxylase (Ddc), which catalyzes conversion of L-5-hydroxytryptophan to 5-HT. DISCUSSION: Our studies possibly suggest a new paradigm relating to how 5-HT operates in the placenta, namely as a factor regulating metabolic functions and blood coagulation. We further suggest that pTGC might be functional analogs of enterochromaffin 5-HT-positive cells of the intestinal mucosa, which regulate similar activities within the gut. Further work, including proteomics and metabolomic studies, are needed to buttress our hypothesis.


Asunto(s)
Placenta/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Serotonina/fisiología , Animales , Dopa-Decarboxilasa/genética , Dopa-Decarboxilasa/metabolismo , Femenino , Células Gigantes/fisiología , Intestinos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/química , Placenta/citología , Embarazo , ARN/análisis , Análisis de Secuencia de ARN , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Trofoblastos/citología , Regulación hacia Arriba
4.
PLoS One ; 16(2): e0247311, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33606835

RESUMEN

The serotonin transporter (5-HTT) is a key molecule of serotoninergic neurotransmission and target of many anxiolytics and antidepressants. In humans, 5-HTT gene variants resulting in lower expression levels are associated with behavioral traits of anxiety. Furthermore, functional magnetic resonance imaging (fMRI) studies reported increased cerebral blood flow (CBF) during resting state (RS) and amygdala hyperreactivity. 5-HTT deficient mice as an established animal model for anxiety disorders seem to be well suited for investigating amygdala (re-)activity in an fMRI study. We investigated wildtype (5-HTT+/+), heterozygous (5-HTT+/-), and homozygous 5-HTT-knockout mice (5-HTT-/-) of both sexes in an ultra-high-field 17.6 Tesla magnetic resonance scanner. CBF was measured with continuous arterial spin labeling during RS, stimulation state (SS; with odor of rats as aversive stimulus), and post-stimulation state (PS). Subsequently, post mortem c-Fos immunohistochemistry elucidated neural activation on cellular level. The results showed that in reaction to the aversive odor CBF in total brain and amygdala of all mice significantly increased. In male 5-HTT+/+ mice amygdala RS CBF levels were found to be significantly lower than in 5-HTT+/- mice. From RS to SS 5-HTT+/+ amygdala perfusion significantly increased compared to both 5-HTT+/- and 5-HTT-/- mice. Perfusion level changes of male mice correlated with the density of c-Fos-immunoreactive cells in the amygdaloid nuclei. In female mice the perfusion was not modulated by the 5-Htt-genotype, but by estrous cycle stages. We conclude that amygdala reactivity is modulated by the 5-Htt genotype in males. In females, gonadal hormones have an impact which might have obscured genotype effects. Furthermore, our results demonstrate experimental support for the tonic model of 5-HTTLPR function.


Asunto(s)
Amígdala del Cerebelo/irrigación sanguínea , Ansiedad/diagnóstico por imagen , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/genética , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Femenino , Hormonas Gonadales/metabolismo , Homocigoto , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Caracteres Sexuales
5.
Artículo en Inglés | MEDLINE | ID: mdl-32800867

RESUMEN

Rats display a rich social behavioral repertoire. An important component of this repertoire is the emission of whistle-like calls in the ultrasonic range, so-called ultrasonic vocalizations (USV). Long low-frequency 22-kHz USV occur in aversive situations, including aggressive interactions, predator exposure, and electric shocks during fear conditioning. They are believed to reflect a negative affective state akin to anxiety and fear. A prominent theory suggests that 22-kHz USV function as alarm calls to warn conspecifics. Serotonin (5-hydroxytryptamine, 5-HT) is strongly implicated in the regulation of affective states, particularly anxiety and fear. A key component of the system is the 5-HT transporter (5-HTT, also known as SERT), regulating 5-HT availability in the synaptic cleft. In the present experiment, we studied the effects of SERT deficiency on overt fear-related behavior and alarm 22-kHz USV during fear conditioning in male and female rats. While overt fear-related behavior was not affected by SERT deficiency and sex, the emission of alarm 22-kHz USV was clearly reduced in homozygous SERT-/- but not heterozygous SERT+/- mutants, as compared to their wildtype SERT+/+ littermate controls. Genotype effects were particularly prominent in females. Females in general emitted fewer alarm 22-kHz USV than males. This supports the view that 22-kHz USV are, at least partly, independently regulated from anxiety or fear and as socially mediated alarm calls do not simply express a negative affective state. Reduced 22-kHz USV emission in rats lacking SERT might be due to social deficits in the use of 22-kHz USV as a socio-affective signal to warn conspecifics about threats.


Asunto(s)
Condicionamiento Psicológico/fisiología , Miedo/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Ondas Ultrasónicas , Vocalización Animal/fisiología , Animales , Miedo/psicología , Femenino , Masculino , Ratas , Ratas Transgénicas , Ratas Wistar , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
6.
J Psychopharmacol ; 34(12): 1408-1417, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33103571

RESUMEN

BACKGROUND: Inhaling 35% carbon dioxide induces an emotional and symptomatic state in humans closely resembling naturally occurring panic attacks, the core symptom of panic disorder. Previous research has suggested a role of the serotonin system in the individual sensitivity to carbon dioxide. In line with this, we previously showed that a variant in the SLC6A4 gene, encoding the serotonin transporter, moderates the fear response to carbon dioxide in humans. To study the etiological basis of carbon dioxide-reactivity and panic attacks in more detail, we recently established a translational mouse model. AIM: The purpose of this study was to investigate whether decreased expression of the serotonin transporter affects the sensitivity to carbon dioxide. METHODS: Based on our previous work, wildtype and serotonin transporter deficient (+/-, -/-) mice were monitored while being exposed to carbon dioxide-enriched air. In wildtype and serotonin transporter +/- mice, also cardio-respiration was assessed. RESULTS: For most behavioral measures under air exposure, wildtype and serotonin transporter +/- mice did not differ, while serotonin transporter -/- mice showed more fear-related behavior. Carbon dioxide exposure evoked a marked increase in fear-related behaviors, independent of genotype, with the exception of time serotonin transporter -/- mice spent in the center zone of the modified open field test and freezing in the two-chamber test. On the physiological level, when inhaling carbon dioxide, the respiratory system was strongly activated and heart rate decreased independent of genotype. CONCLUSION: Carbon dioxide is a robust fear-inducing stimulus. It evokes inhibitory behavioral responses such as decreased exploration and is associated with a clear respiratory profile independent of serotonin transporter genotype.


Asunto(s)
Conducta Animal , Dióxido de Carbono/farmacología , Miedo , Trastorno de Pánico/inducido químicamente , Trastorno de Pánico/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Dióxido de Carbono/administración & dosificación , Modelos Animales de Enfermedad , Miedo/efectos de los fármacos , Miedo/fisiología , Genotipo , Masculino , Ratones , Ratones Noqueados , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia
7.
Neuroimage ; 222: 116975, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32474079

RESUMEN

Life threatening fear after a single exposure evolves in a subset of vulnerable individuals to anxiety, which may persist for their lifetime. Yet neither the whole brain's response to innate acute fear nor how brain activity evolves over time is known. Sustained neuronal activity may be a factor in the development of a persistent fear response. We couple two experimental protocols to provoke acute fear leading to prolonged fear: Predator stress (PS), a naturalistic approach to induce fear in rodents; and Serotonin transporter knockout mouse (SERT-KO) that responds to PS with sustained defensive behavior. Behavior was monitored before, during and at short and long times after PS in wild type (WT) and SERT-KO mice. Both genotypes responded to PS with defensive behavior. SERT-KO retained defensive behavior for 23 days, while WT mice returned to baseline exploratory behavior by 9 days. Thus, differences in neural activity between WT and SERT-KO 9 days after PS identifies neural correlates of persistent defensive behavior, in mice. We used longitudinal manganese-enhanced magnetic resonance imaging (MEMRI) to identify brain-wide neural activity associated with different behaviors. Mn2+ accumulation in active neurons occurs in awake, behaving mice and is retrospectively imaged. Following the same two cohorts of mice, WT and SERT-KO, longitudinally allowed unbiased quantitative comparisons of brain-wide activity by statistical parametric mapping (SPM). During natural behavior in WT, only low levels of activity-induced Mn2+-accumulation were detected, while much more accumulation appeared immediately after PS in both WT and SERT-KO, and evolved at 9 days to a new activity pattern (p < 0.0001, uncorr., T = 5.4). Patterns of accumulation differed between genotypes, with more regions of the brain and larger volumes within regions involved in SERT-KO than WT. A new computational segmentation analysis, using our InVivo Atlas based on a manganese-enhanced MR image of a living mouse, revealed dynamic changes in the volume of significantly enhanced voxels within each segment that differed between genotypes across 45 of 87 segmented regions. At Day 9 after PS, the striatum and ventral pallidum were active in both genotypes but more so in the SERT-KO. SERT-KO also displayed sustained or increased volume of Mn2+ accumulations between Post-Fear and Day 9 in eight segments where activity was decreased or silenced in WT. C-fos staining, an alternative neural activity marker, of brains from the same mice fixed at conclusion of imaging sessions confirmed that MEMRI detected active neurons. Intensity measurements in 12 regions of interest (ROIs) supported the SPM results. Between group comparisons by SPM and of ROI measurements identified specific regions differing between time points and genotypes. We report brain-wide activity in response to a single exposure of acute fear, and, for the first time, its evolution to new activity patterns over time in individuals vulnerable to persistent fear. Our results show multiple regions with dynamic changes in neural activity and that the balance of activity between segments is disordered in the SERT-KO. Thus, longitudinal MEMRI represents a powerful approach to discover how brain-wide activity evolves from the natural state either after an experience or during a disease process.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/fisiología , Miedo/fisiología , Imagen por Resonancia Magnética , Manganeso , Neuroimagen , Estrés Psicológico/fisiopatología , Animales , Encéfalo/diagnóstico por imagen , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/fisiología , Humanos , Aumento de la Imagen , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroimagen/métodos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Estrés Psicológico/diagnóstico por imagen
8.
Cell Physiol Biochem ; 54(1): 126-141, 2020 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-32017483

RESUMEN

BACKGROUND/AIMS: Serotonin (5-hydroxytryptamine, 5-HT) is a neurotransmitter and hormone with important physiological functions in many organs, including the intestine. We have previously shown that 5-HT activates the aryl hydrocarbon receptor (AhR) in intestinal epithelial cells (IECs) via a serotonin transporter (SERT)-dependent mechanism. AhR is a nuclear receptor that binds a variety of molecules including tryptophan (TRP) metabolites to regulate physiological processes in the intestine including xenobiotic detoxification and immune modulation. We hypothesized that 5-HT activates AhR indirectly by interfering with metabolic clearance of AhR ligands by cytochrome P450 1A1 (CYP1A1). METHODS: Inhibition of CYP1A1 activity by 5-HT was assessed in the human intestinal epithelial cell line Caco-2 and recombinant CYP1A1 microsomes using both luciferase and LC-MS/MS. Degradation of 5-HT by recombinant CYP1A1 was measured by LC-MS/MS. For in vitro studies, CYP1A1 and CYP1B1 mRNA expression levels were measured by RT-PCR and CYP1A1 activity was measured by ethoxyresorufin-O-deethylase (EROD) assays. For in vivo studies, AhR ligands were administered to SERT KO mice and WT littermates and intestinal mucosa CYP1A1 mRNA was measured. RESULTS: We show that 5-HT inhibits metabolism of both the pro-luciferin CYP1A1 substrate Luc-CEE as well as the high affinity AhR ligand 6-formylindolo[3,2-b] carbazole (FICZ). Recombinant CYP1A1 assays revealed that 5-HT is metabolized by CYP1A1 in an NADPH dependent manner. Treatment with 5-HT in TRP-free medium, which is devoid of trace AhR ligands, showed that 5-HT requires the presence of AhR ligands to activate AhR. Cotreatment with 5-HT and FICZ confirmed that 5-HT potentiates induction of AhR target genes by AhR ligands. However, this was only true for ligands which are CYP1A1 substrates such as FICZ. Administration of ß-napthoflavone by gavage or indole-3-carbinol via diet to SERT KO mice revealed that lack of SERT impairs intestinal AhR activation. CONCLUSION: Our studies provide novel evidence of crosstalk between serotonergic and AhR signaling where 5-HT can influence the ability of AhR ligands to activate the receptor in the intestine.


Asunto(s)
Citocromo P-450 CYP1A1/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Serotonina/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Células CACO-2 , Carbazoles/farmacología , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1B1/genética , Citocromo P-450 CYP1B1/metabolismo , Humanos , Ligandos , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/metabolismo , Receptores de Hidrocarburo de Aril/química , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Transducción de Señal/efectos de los fármacos , Especificidad por Sustrato , beta-naftoflavona/administración & dosificación
9.
Neuropharmacology ; 168: 107998, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32061666

RESUMEN

Traditional monoaminergic treatments of depression frequently exhibit suboptimal tolerability and effectiveness. The 'short' (s) allele variant of 5-HTTLPR is known to compromise transcriptional efficacy of the serotonin transporter (5-HTT) and can reduce treatment response to traditional antidepressants (e.g. selective serotonin reuptake inhibitors or SSRIs). This study sought to establish the 5-HTT knock-out (KO) line as a mouse model of SSRI-resistant depression and assess its response to a novel glutamatergic antidepressant, ketamine, a non-competitive N-methyl-d-aspartate receptor (NMDAR) antagonist. Following acute antidepressant treatment, 5-HTT KO mice and wild-type (WT) controls were subjected to the forced-swim test (FST), one of the most widely used techniques to detect acute antidepressant response. As hypothesised, when assessed 30 min after administration in the FST, the SSRI sertraline (20 mg/kg, i.p.) produced antidepressant-like effects in WT control but not in 5-HTT KO mice. In contrast, ketamine (20 mg/kg, i.p.) induced antidepressant-like effects in both genotypes. 5-HTT KO mice also exhibited a reduced locomotor response to both MK-801 (another NMDAR antagonist) and ketamine, and reduced GluN2A protein levels in the hippocampus, suggesting glutamatergic dysfunction in this model. These results highlight the utility of 5-HTT KO mice as a relevant model of SSRI-resistant depression and demonstrate that ketamine can produce acute antidepressant-like effects in conditions of 5-HTT deficiency. These findings extend existing literature that indicates ketamine is effective in ameliorating symptoms of treatment-resistant depression and may have implications for understanding the cellular and molecular mechanisms underlying the antidepressant effects of ketamine. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Depresión/metabolismo , Modelos Animales de Enfermedad , Ketamina/uso terapéutico , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Animales , Antidepresivos/farmacología , Depresión/genética , Maleato de Dizocilpina , Femenino , Ketamina/farmacología , Locomoción/efectos de los fármacos , Locomoción/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
10.
Behav Brain Res ; 379: 112387, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31783087

RESUMEN

As the first-line antidepressant drugs, selective serotonin reuptake inhibitors (SSRIs) have efficacy in controlling the symptoms of depression. However, adverse events such as anxiety and hearing disorders were usually observed in patients and even healthy volunteers during the initial phase of SSRI administration. Hearing disorders, including auditory hallucination and tinnitus, are not only highly comorbid with mental disorders but also acknowledged factors that induce psychiatric disorders. The pharmacological and neural mechanisms underlying SSRI-induced anxiety and hearing disorders are not clear. In particularly, the methods evaluating hearing disorders are not well established in animal models, limiting the pre-clinical research on its mechanism. In the present study, we examined the mismatch negativity (MMN), a cognitive component of auditory event-related potential (ERP), to evaluate the hearing process of auditory cortex in mice. Under the acute administration of citalopram, a widely used SSRI, the anxiety-related behaviors and reduced MMN were observed in mice. Serotonin transporter (SERT) is a potential target of SSRIs. The anxiety-related behaviors and reduced MMN were also observed in SERT knockout mice, implying the role of SERT in anxiety and hearing disorders induced by SSRIs. Meanwhile, the auditory brainstem response and initial components of auditory ERP were kept intact in SERT knockout mice, suggesting that hearing neural pathway is less affected by serotonergic system. Our study suggests that the SERT deficient mice might represent a useful animal model in the investigation of the anxiety and hearing disorders during the SSRI treatment.


Asunto(s)
Ansiedad/inducido químicamente , Corteza Auditiva/efectos de los fármacos , Citalopram/efectos adversos , Potenciales Evocados Auditivos/efectos de los fármacos , Trastornos de la Audición/inducido químicamente , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Drug Alcohol Depend ; 205: 107610, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31606593

RESUMEN

BACKGROUND: Human neuroimaging studies indicate that the amygdala plays a key role in cocaine addiction. One key plasticity factor that modulates effects of cocaine on the brain is Brain-Derived Neurotrophic Factor (BDNF). A wealth of evidence shows that cocaine exposure alters BDNF signaling in corticolimbic structures, but, surprisingly, such evidence is very limited for the amygdala. Additionally, while BDNF is strongly regulated by serotonin levels and inherited serotonin transporter down-regulation is associated with increased vulnerability to cocaine addiction, the effects of serotonin transporter genotype on BDNF signaling in the amygdala under naïve and cocaine exposure conditions are unknown. METHODS: We measured BDNF signaling in the central amygdala of wild-type and serotonin transporter knockout rats 24 h into withdrawal from long-access cocaine self-administration. RESULTS: In wild-type rats mature BDNF (mBDNF) protein levels were decreased, whereas the phosphorylation of its receptor TrkB as well as of its intracellular signaling molecules Akt and ERK1 were increased. mBDNF protein expression and its signaling in cocaine-naïve serotonin transporter knockout rats resembled that of wild-type rats with a history of long-access cocaine self-administration. Interestingly, cocaine-exposed serotonin transporter knockout rats showed increased BDNF levels, with no signs of phospho-TrkB receptor coupling to phospho-Akt and phospho-ERK1. CONCLUSIONS: Long-access cocaine self-administration dysregulates BDNF signaling in the central amygdala. Vulnerability to cocaine addiction is associated with dysregulation of this signaling.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Núcleo Amigdalino Central/metabolismo , Cocaína/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Transducción de Señal/fisiología , Animales , Núcleo Amigdalino Central/efectos de los fármacos , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/metabolismo , Masculino , Ratas , Ratas Wistar , Receptor trkB/metabolismo , Autoadministración , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Transducción de Señal/efectos de los fármacos
12.
Neuropsychopharmacol Rep ; 39(2): 130-133, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30719871

RESUMEN

AIM: Repeated psychostimulant drug treatment, including methamphetamine, in rodents readily produces behavioral sensitization, which reflects altered brain function caused by repeated drug exposure. Dendritic remodeling of medium spiny neurons in the nucleus accumbens is thought to be an essential mechanism underlying behavioral sensitization. We recently showed that chronic methamphetamine treatment did not produce behavioral sensitization in serotonin transporter knockout mice. METHODS: In this study, we report the spine density of medium spiny neurons in the nucleus accumbens after repeated methamphetamine injection to examine morphological alterations in serotonin transporter knockout mice. RESULTS: Golgi-COX staining clearly showed that the spine density of medium spiny neurons in the nucleus accumbens increased following repeated methamphetamine treatment in both wild-type and serotonin transporter knockout mice. CONCLUSIONS: Our results suggested that augmented serotonergic neurotransmission produced by serotonin transporter deletion prevents the development of behavioral sensitization in a manner that is independent of dendritic remodeling in the nucleus accumbens.


Asunto(s)
Estimulantes del Sistema Nervioso Central/farmacología , Espinas Dendríticas/efectos de los fármacos , Metanfetamina/farmacología , Núcleo Accumbens/efectos de los fármacos , Animales , Espinas Dendríticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/citología , Núcleo Accumbens/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
13.
Sci Rep ; 9(1): 20283, 2019 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-31889084

RESUMEN

It has been widely established that serotonin plays important role in the regulation of emotional and social behaviour. Rodents with a genetic deletion of the serotonin reuptake transporter (SERT) are used as a model to study lifelong consequences of increased extracellular 5-HT levels due to its impaired reuptake. SERT knock-out (SERT-KO) mice and rats consistently showed anxiety-like symptoms and social deficits. Nevertheless, the impact of SERT deletion on socioemotional ultrasonic communication has not been addressed. Here we investigated the impact of lifelong serotonin abundance on ultrasonic vocalisation accompanying social interactions and open field exploration in rats. SERT-KO rats displayed reduced overall duration of social contacts, but increased time spent on following the conspecific. The altered pattern of social behaviour in SERT-KO rats was accompanied by the structural changes in ultrasonic vocalisations, as they differed from their controls in distribution of call categories. Moreover, SERT deletion resulted in anxiety-like behaviours assessed in the open field test. Their anxious phenotype resulted in a lower tendency to emit appetitive 50-kHz calls during novelty exploration. The present study demonstrates that genetic deletion of SERT not only leads to the deficits in social interaction and increased anxiety but also affects ultrasonic communication.


Asunto(s)
Conducta Animal , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Conducta Social , Ondas Ultrasónicas , Vocalización Animal , Animales , Técnicas de Inactivación de Genes , Masculino , Aprendizaje por Laberinto , Ratas , Ratas Transgénicas
14.
Gigascience ; 7(12)2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30418576

RESUMEN

Background: Active sensing is crucial for navigation. It is characterized by self-generated motor action controlling the accessibility and processing of sensory information. In rodents, active sensing is commonly studied in the whisker system. As rats and mice modulate their whisking contextually, they employ frequency and amplitude modulation. Understanding the development, mechanisms, and plasticity of adaptive motor control will require precise behavioral measurements of whisker position. Findings: Advances in high-speed videography and analytical methods now permit collection and systematic analysis of large datasets. Here, we provide 6,642 videos as freely moving juvenile (third to fourth postnatal week) and adult rodents explore a stationary object on the gap-crossing task. The dataset includes sensory exploration with single- or multi-whiskers in wild-type animals, serotonin transporter knockout rats, rats received pharmacological intervention targeting serotonergic signaling. The dataset includes varying background illumination conditions and signal-to-noise ratios (SNRs), ranging from homogenous/high contrast to non-homogenous/low contrast. A subset of videos has been whisker and nose tracked and are provided as reference for image processing algorithms. Conclusions: The recorded behavioral data can be directly used to study development of sensorimotor computation, top-down mechanisms that control sensory navigation and whisker position, and cross-species comparison of active sensing. It could also help to address contextual modulation of active sensing during touch-induced whisking in head-fixed vs freely behaving animals. Finally, it provides the necessary data for machine learning approaches for automated analysis of sensory and motion parameters across a wide variety of signal-to-noise ratios with accompanying human observer-determined ground-truth.


Asunto(s)
Conducta Animal/fisiología , Bases de Datos Factuales , Grabación en Video , Algoritmos , Animales , Interpretación de Imagen Asistida por Computador , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Transgénicas , Ratas Wistar , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Relación Señal-Ruido , Vibrisas/fisiología
15.
Rev. Hosp. Ital. B. Aires (2004) ; 38(1): 5-10, mar. 2018. tab.
Artículo en Inglés | LILACS | ID: biblio-1023455

RESUMEN

The dysfunction in the serotoninergic neurotransmission has been classically associated with major depressive disorder (MDD); however, other pathways and processes seem to have a role in this illness, such as neurogenesis and related molecules: the Brain-Derived Neurotrophic Factor (BDNF) and the Apolipoprotein E (APOE). There are many reports that indicate an association between certain polymorphism in these genes and MDD. The aim of our study was to analyze the possible association between MDD and polymorphisms in HTR2A (5-hydroxytryptamine receptor 2A), BDNF and APOE genes in a sample of the Argentinean population previously studied for 2 polymorphisms in SLC6A4 (Solute Carrier Family 6 Member 4) gene. Five polymorphisms were studied (rs6311 and rs6313 in HTR2A; rs429358 and rs7412 in APOE, and rs6265 in BDNF) in 95 MDD patients and 107 non-related controls. No statistically significant differences were observed between groups when analyzing the association with a single marker using logistic regression; however, when a possible combinatory effect of the polymorphisms (including previously studied polymorphisms in SLC6A4 gene) was analyzed using a dominant model for the risk alleles, the genotypes L/S_10/12_G/A (OR=3.57(95%CI=1.43-8.93); p=0.004, adjusted p-value=0.01) in SLC6A4 and BDNF genes and L/S_10/12_T/C_3/3_G/A in SLC6A4, HTR2A, APOE and BDNF genes (OR=5.99(95%CI=1.66-21.56); p=0.002, adjusted p-value=0.07), were more prevalent in patients than in controls (20%vs.6% and 15%vs.3%, respectively). Even though it is necessary to replicate these findings in a larger population, our results suggest a possible interaction between molecules involved in neurogenesis (BDNF and APOE), serotoninergic neurotransmission (SLC6A4 and HTR2A) and the pathogenesis of MDD. (AU)


La disfunción en la neurotransmisión serotoninérgica ha sido clásicamente asociada con el trastorno depresivo mayor (TDM); sin embargo, otras vías y procesos parecerían tener un rol en esta enfermedad, como la neurogénesis y moléculas asociadas: el factor neurotrófico derivado del cerebro (BDNF) y la apoliproteína E (APOE). Existen reportes en los que se establecen asociaciones entre polimorfismos en estos genes y el TDM. El objetivo de nuestro trabajo fue analizar la posible asociación entre el TDM y polimorfismos en los genes HTR2A (receptor 5-hidroxitriptamina 2A), BDNF y APOE en una muestra de la población argentina previamente estudiada para 2 polimorfismos en el gen SLC6A4 (transportador soluble familia 6 miembro 4). Se estudiaron 5 polimorfismos (rs6311 y rs6313 en HTR2A; rs429358 y rs7412 en APOE; rs6265 en BDNF) en 95 pacientes con TDM y 107 controles no relacionados. No se observaron diferencias significativas entre grupos al analizar la asociación por regresión logística con un único marcador; cuando se analizó el posible efecto combinatorio de polimorfismos (incluyendo los previamente estudiados para el gen SCL6A4) usando un modelo dominante para los alelos de riesgo, los genotipos L/S_10/12_G/A (OR=3,57(95%CI=1,43-8,93); p=0,004, valor-p-ajustado=0,01) en SLC6A4 y BDNF y L/S_10/12_T/C_3/3_G/A en SLC6A4, HTR2A, APOE y BDNF (OR=5,99(95%CI=1,66-21,56); p=0,002, valor-p-ajustado=0,07), fueron más prevalentes en pacientes que controles (20%vs.6% y 15%vs.3% respectivamente). Si bien es necesario replicar estos hallazgos en una población más grande, nuestros resultados sugieren una posible interacción entre moléculas involucradas en la neurogénesis (BDNF y APOE), la neurotransmisión serotoninérgica (SLC6A4 y HTR2A) y la patogenia de la depresión mayor. (AU)


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Adulto Joven , Apolipoproteínas E/deficiencia , Polimorfismo Genético , Factor Neurotrófico Derivado del Encéfalo/deficiencia , Receptores de Serotonina 5-HT2/deficiencia , Trastorno Depresivo Mayor/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Apolipoproteínas E/genética , Argentina/epidemiología , Factor Neurotrófico Derivado del Encéfalo/genética , Receptores de Serotonina 5-HT2/genética , Trastorno Depresivo Mayor/patología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
16.
CNS Neurosci Ther ; 24(9): 810-819, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29427306

RESUMEN

AIMS: One potential risk factor for posttraumatic stress disorder (PTSD) involves the low activity (short; s) allelic variant of the serotonin transporter-linked polymorphic region (5-HTTLPR), possibly due to reduced prefrontal control over the amygdala. Evidence shows that DNA methylation/demethylation is crucial for fear extinction in these brain areas and is associated with neuronal activation marker c-Fos expression. We hypothesized that impaired fear extinction in serotonin transporter knockout (5-HTT-/- ) rats is related to changes in DNA (de) methylation and c-Fos expression in the prefrontal cortex (PFC) and/or amygdala. METHODS: 5-HTT-/- and 5-HTT+/+ rats were subjected to fear extinction. 2 hours after the extinction session, the overall levels of DNA methylation (5-mC), demethylation (5-hmC), and c-Fos in fear extinction and nonfear extinction rats were measured by immunohistochemistry. RESULTS: 5-HTT-/- rats displayed decreased fear extinction. This was associated with reduced c-Fos activity in the infralimbic PFC. In the central nucleus of the amygdala, c-Fos immunoreactivity was increased in the fear extinction group compared to the no-fear extinction group, regardless of genotype. 5-hmC levels were unaltered in the PFC, but reduced in the amygdala of nonextinction 5-HTT-/- rats compared to nonextinction wild-type rats, which caught up to wild-type levels during fear extinction. 5-mC levels were stable in central amygdala in both wild-type and 5-HTT-/- extinction rats. Finally, c-Fos and 5-mC levels were correlated with the prelimbic PFC, but not amygdala. CONCLUSIONS: In the amygdala, DNA demethylation, independent from c-Fos activation, may contribute to individual differences in risk for PTSD, as conferred by the 5-HTTLPR s-allele.


Asunto(s)
5-Metilcitosina/análogos & derivados , Amígdala del Cerebelo/metabolismo , Extinción Psicológica/fisiología , Miedo/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , 5-Metilcitosina/biosíntesis , Animales , Metilación de ADN , Miedo/psicología , Masculino , Ratas , Ratas Wistar , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
17.
Behav Brain Res ; 346: 16-20, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29233642

RESUMEN

Life stress increases risk for developing post-traumatic stress disorder (PTSD), and more prominently so in short-allele carriers of the serotonin transporter linked polymorphic region (5-HTTLPR). Serotonin transporter knockout (5-HTT-/-) rats show compromised extinction (recall) of conditioned fear, which might mediate the increased risk for PTSD and reduce the therapeutic efficacy of exposure therapy. Here, we assessed whether acute inescapable stress (IS) differentially affects fear extinction and extinction recall in 5-HTT-/- rats and wildtype controls. Surprisingly, IS experience improved fear extinction recall in 5-HTT-/- rats to the level of wildtype animals, while wildtypes were unaffected by this IS. Thus, whereas 5-HTT-/- rats evidently were more responsive to the stressor, the behavioral consequences presented themselves as adaptive.


Asunto(s)
Extinción Psicológica/fisiología , Miedo/fisiología , Recuerdo Mental/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Estrés Psicológico/metabolismo , Análisis de Varianza , Animales , Condicionamiento Psicológico/fisiología , Electrochoque , Reacción Cataléptica de Congelación/fisiología , Masculino , Pruebas Psicológicas , Ratas Transgénicas , Ratas Wistar , Resiliencia Psicológica , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
18.
Neuroscience ; 371: 407-419, 2018 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-29288796

RESUMEN

Autism Spectrum Disorder (ASD) is characterized by impairments in social interaction, social communication, and repetitive and stereotyped behaviors. Recent work has begun to explore gene × environmental interactions in the etiology of ASD. We previously reported that prenatal stress exposure in stress-susceptible heterozygous serotonin transporter (SERT) KO pregnant dams in a mouse model resulted in autism-like behavior in the offspring (SERT/S mice). The association between prenatal stress and ASD appears to be affected by maternal SERT genotype in clinical populations as well. Using the mouse model, we examined autistic-like behaviors in greater detail, and additionally explored whether diet supplementation with docosahexaenoic acid (DHA) may mitigate the behavioral changes. Only male SERT/S mice showed social impairment and stereotyped behavior, and DHA supplementation ameliorated some of these behaviors. We also measured monoamine levels in the SERT/S mice after three treatment paradigms: DHA-rich diet continuously from breeding (DHA diet), DHA-rich diet only after weaning (CTL/DHA diet) and control diet only (CTL diet). The dopamine (DA) content in the striatum was significantly increased in the SERT/S mice compared with wild-type (WT) mice, whereas no difference was observed with noradrenaline and serotonin content. Moreover, DA content in the striatum was significantly reduced in the SERT/S mice with the DHA-rich diet provided continuously from breeding. The results indicate that autism-associated behaviors and changes in the dopaminergic system in this setting can be mitigated with DHA supplementation.


Asunto(s)
Trastorno del Espectro Autista/dietoterapia , Cuerpo Estriado/metabolismo , Ácidos Docosahexaenoicos/administración & dosificación , Dopamina/metabolismo , Efectos Tardíos de la Exposición Prenatal , Estrés Psicológico , Animales , Trastorno del Espectro Autista/etiología , Trastorno del Espectro Autista/metabolismo , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Norepinefrina/metabolismo , Embarazo , Complicaciones del Embarazo/fisiopatología , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Estrés Psicológico/fisiopatología
19.
Nature ; 550(7674): S2-S3, 2017 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-28976951

Asunto(s)
Sexología/tendencias , Conducta Sexual/fisiología , Conducta Sexual/psicología , Salud de la Mujer/tendencias , Animales , Antidepresivos/uso terapéutico , Nivel de Alerta/efectos de los fármacos , Nivel de Alerta/fisiología , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Terapia Cognitivo-Conductual , Anticonceptivos Orales/efectos adversos , Dopamina/metabolismo , Aprobación de Drogas , Epinefrina/metabolismo , Femenino , Humanos , Hipotiroidismo/complicaciones , Hipotiroidismo/tratamiento farmacológico , Libido/efectos de los fármacos , Libido/fisiología , Masculino , Melanocortinas/metabolismo , Orgasmo/fisiología , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Calidad de Vida , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Globulina de Unión a Hormona Sexual/metabolismo , Conducta Sexual/efectos de los fármacos , Disfunciones Sexuales Psicológicas/complicaciones , Disfunciones Sexuales Psicológicas/fisiopatología , Disfunciones Sexuales Psicológicas/psicología , Disfunciones Sexuales Psicológicas/terapia , Tabú/psicología , Estados Unidos , United States Food and Drug Administration , Vagina/anatomía & histología , Vagina/irrigación sanguínea , Vagina/fisiología , alfa-MSH/farmacología , alfa-MSH/uso terapéutico
20.
Behav Brain Res ; 332: 355-361, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28629963

RESUMEN

Along with being the main target of many antidepressant medications, the serotonin transporter (5-HTT) is known to be involved in the pathophysiology of depression and anxiety disorders. In line with this, mice with varying 5-HTT genotypes are invaluable tools to study depression- and anxiety-like behaviours as well as the mechanisms mediating potential therapeutics. There is clear evidence that both genetic and environmental factors play a role in the aetiology of psychiatric disorders. In that regard, housing paradigms which seek to enhance cognitive stimulation and physical activity have been shown to exert beneficial effects in animal models of neuropsychiatric disorders. In the present study, we examined the effects of environmental enrichment on affective-like behaviours and sensorimotor gating function of 5-HTT knock-out (KO) mice. Using the elevated-plus maze and the light-dark box, we found that environmental enrichment ameliorated the abnormal innate anxiety of 5-HTT KO mice on both tests. In contrast, environmental enrichment did not rescue the depression-like behaviour displayed by 5-HTT KO mice in the forced-swim test. Finally, measuring pre-pulse inhibition, we found no effect of genotype or treatment on sensorimotor gating. In conclusion, our data suggest that environmental enrichment specifically reduces innate anxiety of 5-HTT KO mice with no amelioration of the depression-like behaviour. This has implications for the current use of clinical interventions for patients with symptoms of both anxiety and depression.


Asunto(s)
Ansiedad/metabolismo , Ansiedad/terapia , Depresión/metabolismo , Depresión/terapia , Ambiente , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Afecto/fisiología , Análisis de Varianza , Animales , Conducta Animal/fisiología , Femenino , Vivienda para Animales , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Distribución Aleatoria , Filtrado Sensorial/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA