Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
1.
Transl Psychiatry ; 12(1): 274, 2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35821204

RESUMEN

The serotonin-transporter-linked promoter region (5-HTTLPR) has been widely investigated as contributing to depression vulnerability. Nevertheless, empirical research provides wide contrasting findings regarding its involvement in the etiopathogenesis of the disorder. Our hypothesis was that such discrepancy can be explained considering time as moderating factor. We explored this hypothesis, exploiting a meta analytic approach. We searched PubMed, PsychoINFO, Scopus and EMBASE databases and 1096 studies were identified and screened, resulting in 22 studies to be included in the meta-analyses. The effect of the 5-HTTLPR x stress interaction on depression risk was found to be moderated by the following temporal factors: the duration of stress (i.e. chronic vs. acute) and the time interval between end of stress and assessment of depression (i.e. within 1 year vs. more than 1 year). When stratifying for the duration of stress, the effect of the 5-HTTLPR x stress interaction emerged only in the case of chronic stress, with a significant subgroup difference (p = 0.004). The stratification according to time interval revealed a significant interaction only for intervals within 1 year, though no difference between subgroups was found. The critical role of time interval clearly emerged when considering only chronic stress: a significant effect of the 5-HTTLPR and stress interaction was confirmed exclusively within 1 year and a significant subgroup difference was found (p = 0.01). These results show that the 5-HTTLPR x stress interaction is a dynamic process, producing different effects at different time points, and indirectly confirm that s-allele carriers are both at higher risk and more capable to recover from depression. Overall, these findings expand the current view of the interplay between 5-HTTLPR and stress adding the temporal dimension, that results in a three-way interaction: gene x environment x time.


Asunto(s)
Depresión/etiología , Interacción Gen-Ambiente , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Estrés Psicológico/complicaciones , Depresión/epidemiología , Depresión/genética , Genotipo , Factores de Riesgo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Estrés Psicológico/genética , Factores de Tiempo
2.
PLoS Genet ; 17(11): e1009885, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34735454

RESUMEN

Molecular insights into the selective vulnerability of retinal ganglion cells (RGCs) in optic neuropathies and after ocular trauma can lead to the development of novel therapeutic strategies aimed at preserving RGCs. However, little is known about what molecular contexts determine RGC susceptibility. In this study, we show the molecular mechanisms underlying the regional differential vulnerability of RGCs after optic nerve injury. We identified RGCs in the mouse peripheral ventrotemporal (VT) retina as the earliest population of RGCs susceptible to optic nerve injury. Mechanistically, the serotonin transporter (SERT) is upregulated on VT axons after injury. Utilizing SERT-deficient mice, loss of SERT attenuated VT RGC death and led to robust retinal axon regeneration. Integrin ß3, a factor mediating SERT-induced functions in other systems, is also upregulated in RGCs and axons after injury, and loss of integrin ß3 led to VT RGC protection and axon regeneration. Finally, RNA sequencing analyses revealed that loss of SERT significantly altered molecular signatures in the VT retina after optic nerve injury, including expression of the transmembrane protein, Gpnmb. GPNMB is rapidly downregulated in wild-type, but not SERT- or integrin ß3-deficient VT RGCs after injury, and maintaining expression of GPNMB in RGCs via AAV2 viruses even after injury promoted VT RGC survival and axon regeneration. Taken together, our findings demonstrate that the SERT-integrin ß3-GPNMB molecular axis mediates selective RGC vulnerability and axon regeneration after optic nerve injury.


Asunto(s)
Axones , Regeneración Nerviosa , Enfermedades del Sistema Nervioso/metabolismo , Células Ganglionares de la Retina/citología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/patología
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.
FASEB J ; 35(7): e21701, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34143529

RESUMEN

Irritable bowel syndrome (IBS) is a common functional gastrointestinal disease. Although visceral hypersensitivity (VH) and disturbed gastrointestinal motility are typical pathophysiological features of IBS, the pathological mechanisms underlying this disease remain unclear. Serotonin system abnormalities are considered to play an important role in the pathomechanisms of IBS. Here, we hypothesize that similar alterations, including VH and colonic motility, induced by serotonin transporter (SERT) knockout result from altered serotonin signaling. We sought to determine the molecular mechanism underlying VH and colonic dysmotility induced by SERT knockout. We found that female SERT (slc6a4)-knockout (KO; ie, slc6a4-/- ) rats exhibited lower pain pressure thresholds (PPTs) than wild-type (WT; ie, slc6a4+/+ ) rats in response to colorectal distension (CRD). Significantly increased fecal pellet output and reduced concentration of serum tryptophan were observed in the female SERT KO rats. The concentrations of 5-hydroxytryptamine (5-HT) in platelet-rich plasma (PRP) and serum in SERT KO rats were lower than those in WT rats, but the numbers of enterochromaffin cells (ECs) and the concentrations of 5-HT in colon of SERT KO rats were higher than those of WT rats. Finally, increased expression levels of 5-HT1B receptors, 5-HT2C receptors, 5-HT3A receptors, 5-HT3B receptors, 5-HT6 receptors, 5-HT7 receptors, and glycosylated dopamine transporters (DATs) were found in the female SERT KO rats. We concluded that alterations in the serotonin system induced by the knockout of slc6a4 might result in VH and accelerated gastrointestinal motility in female SERT KO rats, which can be used as an animal model of IBS.


Asunto(s)
Colon/patología , Motilidad Gastrointestinal , Hipersensibilidad/patología , Síndrome del Colon Irritable/patología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Serotonina/metabolismo , Animales , Animales Modificados Genéticamente , Colon/metabolismo , Modelos Animales de Enfermedad , Femenino , Hipersensibilidad/etiología , Hipersensibilidad/metabolismo , Síndrome del Colon Irritable/etiología , Síndrome del Colon Irritable/metabolismo , Ratas , Ratas Sprague-Dawley
5.
J Pharmacol Sci ; 145(4): 297-307, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33712280

RESUMEN

Herein, we investigated the functional association of the serotonin transporter (SERT) with syntaxin-3 (STX3). We first overexpressed SERT and STX3 in various cells and examined their interaction, localization, and functional association. Immunoprecipitation studies revealed that STX3 interacted with SERT when expressed in COS-7 cells. Immunocytochemical studies revealed that SERT and STX3 were colocalized in the endoplasmic reticulum (ER) and Golgi apparatus. STX3 overexpression significantly reduced the uptake activity of SERT by attenuating its plasma membrane expression, suggesting that overexpressed STX3 anchors SERT in the ER and Golgi apparatus. STX3 knockdown did not affect the uptake activity of SERT but altered its glycosylation state. To elucidate the association of STX3 with SERT under physiological conditions, rather than overexpressing cells, we investigated this interaction in polarized Caco-2 cells, which endogenously express both proteins. Immunocytochemical studies revealed that SERT and STX3 were localized in microvilli-like structures at the apical plasma membrane. STX3 knockdown marginally but significantly decreased the serotonin uptake activity of Caco-2 cells, suggesting that STX3 positively regulates SERT function in Caco-2 cells, as opposed to SERT regulation by STX3 in overexpressing cells. Collectively, STX3 may colocalize with SERT during SERT membrane trafficking and regulate SERT function in an STX3-expressing lesion-dependent manner.


Asunto(s)
Epistasis Genética/genética , Expresión Génica/genética , Proteínas Qa-SNARE/metabolismo , Proteínas Qa-SNARE/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Glicosilación , Aparato de Golgi/metabolismo , Microvellosidades/metabolismo , Proteínas Qa-SNARE/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
6.
Neurogastroenterol Motil ; 33(4): e14036, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33222337

RESUMEN

BACKGROUND: Oxidized phospholipid derivatives (OxPAPCs) act as bacterial lipopolysaccharide (LPS)-like damage-associated molecular patterns. OxPAPCs dose-dependently exert pro- or anti-inflammatory effects by interacting with several cellular receptors, mainly Toll-like receptors 2 and 4. It is currently unknown whether OxPAPCs may affect enteric nervous system (ENS) functional and structural integrity. METHODS: Juvenile (3 weeks old) male C57Bl/6 mice were treated intraperitoneally with OxPAPCs, twice daily for 3 days. Changes in small intestinal contractility were evaluated by isometric neuromuscular responses to receptor and non-receptor-mediated stimuli. Alterations in ENS integrity and serotonergic pathways were assessed by real-time PCR and confocal immunofluorescence microscopy in longitudinal muscle-myenteric plexus whole-mount preparations (LMMPs). Tissue levels of serotonin (5-HT), tryptophan, and kynurenine were measured by HPLC coupled to UV/fluorescent detection. KEY RESULTS: OxPAPC treatment induced enteric gliosis, loss of myenteric plexus neurons, and excitatory hypercontractility, and reduced nitrergic neurotransmission with no changes in nNOS+ neurons. Interestingly, these changes were associated with a higher functional response to 5-HT, altered immunoreactivity of 5-HT receptors and serotonin transporter (SERT) together with a marked decrease in 5-HT levels, shifting tryptophan metabolism toward kynurenine production. CONCLUSIONS AND INFERENCES: OxPAPC treatment disrupted structural and functional integrity of the ENS, affecting serotoninergic tone and 5-HT tissue levels toward a higher kynurenine content during adolescence, suggesting that changes in intestinal lipid metabolism toward oxidation can affect serotoninergic pathways, potentially increasing the risk of developing functional gastrointestinal disorders during critical stages of development.


Asunto(s)
Sistema Nervioso Entérico/fisiología , Intestino Delgado/fisiología , Fosfatidilcolinas/farmacología , Receptores de Serotonina/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Serotonina/fisiología , Factores de Edad , Animales , Relación Dosis-Respuesta a Droga , Sistema Nervioso Entérico/efectos de los fármacos , Motilidad Gastrointestinal/efectos de los fármacos , Motilidad Gastrointestinal/fisiología , Intestino Delgado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
7.
Biol Res Nurs ; 22(1): 5-12, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31409118

RESUMEN

Low birth weight is an ongoing public health problem with severe consequences for those affected, including early morbidity and mortality and elevated risk for lifelong deficits in cognitive function. These deficits can be ameliorated by early intervention in many cases. To contribute to criteria for earlier identification of at-risk children prior to the onset of delays or deficits, we examined relationships between three gene candidates-SLC6A4, BDNF, COMT-and cognitive outcomes at school age in a secondary analysis of existing data from a nationally representative cohort. Single nucleotide polymorphism rs4074134, a variant of BDNF, and a rare insertion/deletion in the intron region of SLC6A4 were significant predictors of cognitive performance. Our final model predicted 17% of the variance in composite cognitive test scores among children with low birth weight at school age (F = 96.36, p < .001, R2 = .17). Specifically, children homozygous for cytosine at rs4074134 scored .62 standard deviations higher on a measure of global cognition than children with one or more thymine. Similarly, children with an extra-long copy number variant of SLC6A4 scored .88 standard deviations higher than children who had one or more short forms of the gene. These findings support the potential for an approach to identifying children with low birth weights who are most at need of early intervention services. Future research should focus on validation of these findings in an independent sample and confirmation of the biological mechanisms through which these genes influence cognitive development.


Asunto(s)
Desarrollo Infantil/fisiología , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/fisiopatología , Predisposición Genética a la Enfermedad , Recién Nacido de Bajo Peso/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Niño , Trastornos del Conocimiento/epidemiología , Estudios de Cohortes , Femenino , Humanos , Recién Nacido , Estudios Longitudinales , Masculino , Factores de Riesgo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Estados Unidos/epidemiología
8.
Med Hypotheses ; 134: 109406, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31634774

RESUMEN

We introduce a novel hypothesis which states that the therapeutic utilisation of psilocybin has beneficial effects on genetic aging. Ex hypothesi, we predict a priori that controlled psilocybin interventions exert quantifiable positive impact on leucocyte telomere length (telomeres are a robust predictor of mortality and multifarious aging-related diseases). Our hypothesising follows the Popperian logic of scientific discovery, viz., bold (and refutable) conjectures form the very foundation of scientific progress. The 'psilocybin-telomere hypothesis' is formalised as a logically valid deductive (syllogistic) argument and we provide substantial evidence to support the underlying premises. Impetus for our theorising derives from a plurality of converging empirical sources indicating that psilocybin has persistent beneficial effects on various aspects of mental health (e.g., in the context of depression, anxiety, PTSD, OCD, addiction, etc.). Additional support is based on a large corpus of studies that establish reliable correlations between mental health and telomere attrition (improved mental health is generally correlated with longer telomeres). Another pertinent component of our argument is based on recent studies which demonstrate that "meditative states of consciousness" provide beneficial effects on genetic aging. Similarly, psilocybin can induce states of consciousness that are neurophysiologically and phenomenologically significantly congruent with meditative states. Furthermore, prior research has demonstrated that a single dose of psilocybin can occasion life-changing transformative experiences (≈ 70% of healthy volunteers rate their experience with psilocybin amongst the five personally most meaningful lifetime events, viz., ranked next to giving birth to a child or losing a loved one). We postulate that these profound psychological events leave quantifiable marks at the molecular genetic/epigenetic level. Given the ubiquitous availability and cost effectiveness of telomere length assays, we suggest that quantitative telomere analysis should be regularly included in future psilocybin studies as an adjunctive biological marker (i.e., to facilitate scientific consilience via methodological triangulation). In order to substantiate the 'psilocybin-telomere hypothesis' potential neuropsychopharmacological, endocrinological, and genetic mechanisms of action are discussed (e.g., HPA-axis reactivity, hippocampal neurogenesis, neurotropic growth factors such as BDNF, 5-HT2A receptor agonism, neuroplasticity/synaptoplasticity, brain-wide alterations in neuronal functional connectivity density, involvement of the SLC6A4 serotonin transporter gene, inter alia). The proposed research agenda is thus intrinsically highly interdisciplinary, and it has deep ramifications from a philosophy of science perspective as it connects the epistemic level (qualitative experiential phenomenology) with the ontic level (quantitative molecular genetics) of analysis. In the long term, multidisciplinary and innovative investigations of the 'psilocybin-telomere hypothesis' could contribute to the improvement of senotherapeutic psychological interventions and the identification of novel geroprotective and neuroprotective/restorative pharmaceutical targets to decelerate genetic aging and improve well-being and quality of life during the aging process.


Asunto(s)
Envejecimiento/efectos de los fármacos , Modelos Genéticos , Modelos Psicológicos , Psilocibina/uso terapéutico , Psicotrópicos/uso terapéutico , Acortamiento del Telómero/efectos de los fármacos , Envejecimiento/genética , Envejecimiento/psicología , Envejecimiento Prematuro/tratamiento farmacológico , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/prevención & control , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/genética , Factor Neurotrófico Derivado del Encéfalo/fisiología , Estado de Conciencia/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Depresión/tratamiento farmacológico , Depresión/genética , Modelos Animales de Enfermedad , Sistema Endocrino/fisiopatología , Humanos , Neurotransmisores/fisiología , Estrés Oxidativo/efectos de los fármacos , Personalidad/efectos de los fármacos , Psilocibina/farmacología , Psicotrópicos/farmacología , Proyectos de Investigación , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/genética , Acortamiento del Telómero/fisiología
9.
Brain Res Bull ; 152: 95-106, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31301378

RESUMEN

Studies in animal models of epilepsy revealed compromised serotonin (5-HT) transmission between the raphe nuclei and the brain limbic system. The goal of the present study was to evaluate the effects of epilepsy on the structural integrity of the dorsal (DR) and median (MnR) raphe nuclei and on the morphology of serotonergic fiber terminals in the dentate gyrus (DG), infralimbic cortex (IL) and medial septum (MS). The study was performed in adult Wistar rats using the kainate (9.5 mg/kg) status epilepticus (SE) model. Four months post-SE, the brainstem sections of the animals were immunostained for 5-HT, whereas the forebrain sections were immunostained for serotonin transporter (SERT). Stereological analysis revealed that epileptic rats, as compared to controls, had approximately 30% less 5-HT-stained cells in the interfascicular part of the DR, but twice as many 5-HT-stained cells in the MnR. Another finding was the reorganization of the 5-HT fiber network in all target areas analyzed, as indicated by the rightward shift of the density-size distribution histograms of SERT-stained fiber varicosities. Nonlinear regression analysis of these histograms revealed that SERT-stained varicosities were represented by two subpopulations characterized by distinct cross-sectional areas. The areal density of the small-sized varicosities was decreased in the DG (hilus and molecular layer), IL cortex (layers II/III) and MS, while that of the larger-sized varicosities was increased. The present results support the hypothesis that chronic epilepsy can trigger profound structural reorganization of the ascending serotonergic pathways in the rat brain.


Asunto(s)
Epilepsia del Lóbulo Temporal/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Serotonina/metabolismo , Animales , Tronco Encefálico/metabolismo , Corteza Cerebral/metabolismo , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Epilepsia/metabolismo , Epilepsia/fisiopatología , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/metabolismo , Ácido Kaínico/farmacología , Masculino , Prosencéfalo/metabolismo , Núcleos del Rafe/metabolismo , Ratas , Ratas Wistar , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología
10.
J Affect Disord ; 256: 205-212, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31181376

RESUMEN

BACKGROUND: Recent meta-analyses stimulate an ongoing debate whether 5-HTTLPR modulates the risk for depression including a more pronounced association between stress and depression in the short (S) allele relative to the long (L) allele. Elucidating the pathways by which 5-HTTLPR contributes to depression could resolve this controversy. Insomnia independently contributes to the onset and course of negative affective symptoms and, hence, represents one of the primary risk factors for depression. To evaluate the relevance of this relationship for the interaction between 5-HTTLPR and stress in depression, the present review investigated the moderating influence of 5-HTTLPR on the relationship between stress and sleep quality as well as on the relationship between sleep and affective symptomatology. METHODS: A systematic search was performed in the PubMed and PsycINFO databases to include a complete outline of studies investigating the relationships of interest. RESULTS: Results of the included articles reveal that the 5-HTTLPR S-allele relative to the L-allele increases the risk for stress-related sleep quality reductions and promotes the negative affective consequences of inadequate sleep. LIMITATIONS: The apparent involvement of sleep in the association between 5-HTTLPR and depression remains to be more directly (empirically) examined and studies exploring the influence of 5-HTTLPR on sleep quality produced inconsistent results. CONCLUSIONS: The reviewed findings support the involvement of sleep in the interaction between 5-HTTLPR and stress in depression. This could have important implications for the inconsistent findings characterizing this field of research and may provide valuable insight into the pathophysiological mechanisms underlying genetic contributions to depression.


Asunto(s)
Depresión/genética , Depresión/fisiopatología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Trastornos del Inicio y del Mantenimiento del Sueño/genética , Sueño/genética , Alelos , Femenino , Genotipo , Humanos , Estudios Longitudinales , Masculino , Factores de Riesgo , Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología
11.
Neuropharmacology ; 161: 107564, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30851308

RESUMEN

The serotonin transporter (SERT) regulates serotonergic neurotransmission by retrieving released serotonin and replenishing vesicular stores. SERT is not only delivered to axons but it is also present on the neuronal soma and on dendrites. It has not been possible to restrict the distribution of SERT without affecting transporter function. Hence, the physiological role of somatodendritic SERT remains enigmatic. The SERT C-terminus harbors a conserved RI-motif, which recruits SEC24C, a cargo receptor in the coatomer protein-II coat. Failure to engage SEC24C precludes axonal enrichment of SERT. Here we introduced a point mutation into the RI-motif of human SERT causing confinement of the resulting - otherwise fully functional - hSERT-R607A on the somatodendritic membrane of primary rat dorsal raphe neurons. Transgenic expression of the corresponding Drosophila mutant dSERT-R599A led to its enrichment in the somatodendritic compartment of serotonergic neurons in the fly brain. We explored the possible physiological role of somatodendritic SERT by comparing flies harboring wild type SERT and dSERT-R599A in a behavioral paradigm for serotonin-modulated odor perception. When globally re-expressed in serotonergic neurons, wild type SERT but not dSERT-R599A restored ethanol preference. In contrast, dSERT-R599A restored ethanol preference after targeted expression in contralaterally projecting, serotonin-immunoreactive deuterocerebral (CSD) interneurons, while expression of wild type SERT caused ethanol aversion. We conclude that, in CSD neurons, (i) somatodendritic SERT supports ethanol attraction, (ii) axonal SERT specifies ethanol aversion, (iii) the effect of axonal SERT can override that of somatodendritic SERT. These observations demonstrate a distinct biological role of somatodendritic and axonal serotonin transport. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.


Asunto(s)
Axones/fisiología , Dendritas/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Olfato/fisiología , Animales , Animales Modificados Genéticamente , Axones/metabolismo , Línea Celular , Depresores del Sistema Nervioso Central/farmacología , Dendritas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Etanol/farmacología , Humanos , Interneuronas/efectos de los fármacos , Mutación Puntual/genética , Cultivo Primario de Células , Pliegue de Proteína , Núcleos del Rafe/citología , Núcleos del Rafe/metabolismo , Ratas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Olfato/genética
12.
Sci Rep ; 9(1): 2138, 2019 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-30765765

RESUMEN

Serotonin transporter (SERT) plays a critical role in regulating extracellular availability of serotonin (5-HT) in the gut and brain. Mice with deletion of SERT develop metabolic syndrome as they age. Changes in the gut microbiota are being increasingly implicated in Metabolic Syndrome and Diabetes. To investigate the relationship between the gut microbiome and SERT, this study assessed the fecal and cecal microbiome profile of 11 to 12 week-old SERT+/+ and SERT-/- mice. Microbial DNA was isolated, processed for metagenomics shotgun sequencing, and taxonomic and functional profiles were assessed. 34 differentially abundant bacterial species were identified between SERT+/+ and SERT-/-. SERT-/- mice displayed higher abundances of Bacilli species including genera Lactobacillus, Streptococcus, Enterococcus, and Listeria. Furthermore, SERT-/- mice exhibited significantly lower abundances of Bifidobacterium species and Akkermansia muciniphilia. Bacterial community structure was altered in SERT-/- mice. Differential abundance of bacteria was correlated with changes in host gene expression. Bifidobacterium and Bacilli species exhibited significant associations with host genes involved in lipid metabolism pathways. Our results show that SERT deletion is associated with dysbiosis similar to that observed in obesity. This study contributes to the understanding as to how changes in gut microbiota are associated with metabolic phenotype seen in SERT deficiency.


Asunto(s)
Bacterias/metabolismo , Disbiosis/metabolismo , Heces/microbiología , Microbioma Gastrointestinal , Regulación de la Expresión Génica , Metagenómica/métodos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Animales , Bacterias/clasificación , Bacterias/genética , Disbiosis/etiología , Disbiosis/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Ribosómico 16S/análisis , ARN Ribosómico 16S/genética
13.
Addict Biol ; 24(3): 344-354, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-29292566

RESUMEN

Counter-conditioning can be a valid strategy to reduce reinstatement of reward-seeking behavior. However, this has not been tested in laboratory animals with extended cocaine-taking backgrounds nor is it well understood, which individual differences may contribute to its effects. Here, we set out to investigate the influence of serotonin transporter (5-HTT) genotype on the effectiveness of counter-conditioning after extended access to cocaine self-administration. To this end, 5-HTT+/+ and 5-HTT-/- rats underwent a touch screen-based approach to test if reward-induced reinstatement of responding to a previously counter-conditioned cue is reduced, compared with a non-counter-conditioned cue, in a within-subject manner. We observed an overall extinction deficit of cocaine-seeking behavior in 5-HTT-/- rats and a resistance to punishment during the counter-conditioning session. Furthermore, we observed a significant decrease in reinstatement to cocaine and sucrose associated cues after counter-conditioning but only in 5-HTT+/+ rats. In short, we conclude that the paradigm we used was able to produce effects of counter-conditioning of sucrose seeking behavior in line with what is described in literature, and we demonstrate that it can be effective even after long-term exposure to cocaine, in a genotype-dependent manner.


Asunto(s)
Cocaína/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Inhibidores de Captación de Dopamina/farmacología , Recompensa , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Análisis de Varianza , Animales , Señales (Psicología) , Extinción Psicológica , Ratas Endogámicas , Refuerzo en Psicología , Autoadministración , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología
14.
Psychother Psychosom Med Psychol ; 69(7): 266-274, 2019 Jul.
Artículo en Alemán | MEDLINE | ID: mdl-30025422

RESUMEN

Post-traumatic stress disorder (PTSD) is a mental disorder following a severe traumatic experience and is characterized by high rates of comorbidity with related psychiatric disorders. However, even for individuals experiencing the same trauma, there is considerable inter-individual variability in the risk of PTSD, and this is largely thought to be determined by biological processes, such as genetic predisposition and epigenetic mechanism. In this review we will summarize recent research on genetics of PTSD, primarily focusing on candidate gene-association studies, targeting on functional genetic variants in the monoaminergic system and the hypothalamic-pituitary-adrenal (HPA) axis. In addition, results from recent genome-wide association studies (GWAS) will be reported and we will highlight the interplay of genetic factors with environmental factors, based on evidence from gene-environment interaction analysis and studies on the epigenetic regulation of PTSD. Finally, we will provide a brief outlook towards the potential and perspectives of pharmaco-genetic studies.


Asunto(s)
Interacción Gen-Ambiente , Trastornos por Estrés Postraumático/genética , Comorbilidad , Enfermedades en Gemelos/diagnóstico , Enfermedades en Gemelos/genética , Enfermedades en Gemelos/fisiopatología , Enfermedades en Gemelos/psicología , Dopamina/fisiología , Estudios de Asociación Genética , Estudio de Asociación del Genoma Completo , Holocausto/psicología , Humanos , Sistema Hipotálamo-Hipofisario/fisiopatología , Individualidad , Farmacogenética , Polimorfismo Genético/genética , Factores de Riesgo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Trastornos por Estrés Postraumático/diagnóstico , Trastornos por Estrés Postraumático/fisiopatología , Trastornos por Estrés Postraumático/psicología , Sobrevivientes/psicología , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
15.
J Pharmacol Sci ; 139(1): 29-36, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30522963

RESUMEN

The serotonin transporter (SERT) is functionally regulated via membrane trafficking. Our previous studies have demonstrated that the SERT C-terminal deletion mutant (SERTΔCT) showed a robust decrease in its membrane trafficking and was retained in the endoplasmic reticulum (ER), suggesting that SERTΔCT is an unfolded protein that may cause ER stress. The Sigma-1 receptor (SigR1) has been reported to attenuate ER stress via its chaperone activity. In this study, we investigated the effects of SKF-10047, a prototype SigR1 agonist, on the membrane trafficking and uptake activity of SERT and SERTΔCT expressed in COS-7 cells. Twenty-four hours of SKF-10047 treatment (>200 µM) accelerated SERT membrane trafficking and robustly upregulated SERTΔCT activity. Interestingly, these effects of SKF-10047 on SERT functions were also found in cells in which SigR1 expression was knocked down by shRNA, suggesting that SKF-10047 exerted these effects on SERT via a mechanism independent of SigR1. A cDNA array study identified several candidate genes involved in the mechanism of action of SKF-10047. Among them, Syntaxin3, a member of the SNARE complex, was significantly upregulated by 48 h of SKF-10047 treatment. These results suggest that SKF-10047 is a candidate for ER stress relief.


Asunto(s)
Membrana Celular/efectos de los fármacos , Fenazocina/análogos & derivados , Receptores sigma/agonistas , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , Estrés del Retículo Endoplásmico , Técnicas de Silenciamiento del Gen , Mutación , Fenazocina/farmacología , Transporte de Proteínas , Receptores sigma/genética , Receptor Sigma-1
16.
Hum Mol Genet ; 28(7): 1153-1161, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30535103

RESUMEN

Elevated serotonin (5-HT) blood levels, the first biomarker identified in autism research, has been consistently found in 20-30% of patients with Autism Spectrum Disorder (ASD). Hyperserotonemia is mainly due to greater 5-HT uptake into platelets, mediated by the 5-HT transporter (SERT) located at the platelet plasma membrane. The protein complex involved in platelet SERT trafficking and externalization includes integrin ß3, the beta subunit of the platelet membrane adhesive GP IIb/IIIa. Integrin ß3 is encoded by the ITGB3 gene, previously identified as a quantitative trait locus (QTL) for 5-HT blood levels in ASD at single nucleotide polymorphism (SNP) rs2317385. The present study aims to identify the functional ITGB3 gene variants contributing to hyperserotonemia. ITGB3 gene sequencing in 20 individuals selected on the basis of rs2317385 genotypes defined four haplotypes encompassing six SNPs located in the ITGB3 gene promoter region, all in linkage disequilibrium with rs2317385. Luciferase assays in two hematopoietic cell lines, K-562 and HEL 92.1.7, demonstrate that ITGB3 gene promoter activity is enhanced by the presence of the C allele at rs55827077 specifically during differentiation into megakaryocytes (P < 0.01), with modulatory effects by flanking SNPs. This same allele is strongly associated with (a) higher 5-HT blood levels in 176 autistic individuals (P < 0.001), (b) greater platelet integrin ß3 protein expression (P < 0.05) and (c) enhanced SERT trafficking from the cytosol toward the platelet plasma membrane (P = 4.05 × 10-11). Our results support rs55827077 as the functional ITGB3 gene promoter variant contributing to elevated 5-HT blood levels in ASD and define a mechanistic chain of events linking ITGB3 to hyperserotonemia.


Asunto(s)
Trastorno del Espectro Autista/genética , Integrina beta3/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Adolescente , Adulto , Trastorno Autístico/genética , Niño , Preescolar , Trastornos de Somnolencia Excesiva/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Genotipo , Haplotipos , Humanos , Integrina beta3/fisiología , Desequilibrio de Ligamiento/genética , Masculino , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Transporte de Proteínas/fisiología , Serotonina/análisis , Serotonina/sangre , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
17.
Mol Psychiatry ; 24(5): 726-745, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30279456

RESUMEN

Antidepressants that block the serotonin transporter, (Slc6a4/SERT), selective serotonin reuptake inhibitors (SSRIs) improve mood in adults but have paradoxical long-term effects when administered during perinatal periods, increasing the risk to develop anxiety and depression. The basis for this developmental effect is not known. Here, we show that during an early postnatal period in mice (P0-P10), Slc6a4/SERT is transiently expressed in a subset of layer 5-6 pyramidal neurons of the prefrontal cortex (PFC). PFC-SERT+ neurons establish glutamatergic synapses with subcortical targets, including the serotonin (5-HT) and GABA neurons of the dorsal raphe nucleus (DRN). PFC-to-DRN circuits develop postnatally, coinciding with the period of PFC Slc6a4/SERT expression. Complete or cortex-specific ablation of SERT increases the number of functional PFC glutamate synapses on both 5-HT and GABA neurons in the DRN. This PFC-to-DRN hyperinnervation is replicated by early-life exposure to the SSRI, fluoxetine (from P2 to P14), that also causes anxiety/depressive-like symptoms. We show that pharmacogenetic manipulation of PFC-SERT+ neuron activity bidirectionally modulates these symptoms, suggesting that PFC hypofunctionality has a causal role in these altered responses to stress. Overall, our data identify specific PFC descending circuits that are targets of antidepressant drugs during development. We demonstrate that developmental expression of SERT in this subset of PFC neurons controls synaptic maturation of PFC-to-DRN circuits, and that remodeling of these circuits in early life modulates behavioral responses to stress in adulthood.


Asunto(s)
Células Piramidales/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Antidepresivos/farmacología , Ansiedad/metabolismo , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/fisiopatología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Depresión/tratamiento farmacológico , Depresión/fisiopatología , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Emociones/efectos de los fármacos , Femenino , Neuronas GABAérgicas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Inhibidores Selectivos de la Recaptación de Serotonina/metabolismo
18.
Behav Brain Res ; 361: 50-53, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30562569

RESUMEN

The serotonin transporter gene (5-HTT) is involved in the regulation of the neural serotonin. Polymorphisms in the 5-HTT gene have been described in many species to be involved in physiological processes and emotions. A functional polymorphism in the 5´-flanking region of the 5-HTT gene is known from chickens, with a deletion-allele (D), which is associated with an increased 5-HTT expression, in comparison to the wild-type-allele (W). In domestic populations, the majority of hens carry the W-allele. The regulatory changes of the 5-HTT are accompanied in chickens, as in humans, by modulations of fear. Beside these effects on fear, the understanding of potential functional consequences on the social behaviour in the gregarious chicken is lacking. Thus, we here investigated whether the 5-HTT polymorphism with three genotypes (WW, WD, DD), is not only linked to fear-related behaviour, but affects also socio-positive and -negative behaviours of adult hens. Our data confirmed the effects on fear-related behaviour. WW hens showed highest levels of fear. Interestingly, no differences in the social behaviours were present between the hens of the different 5-HTT genotypes. We further discuss implications for potential evolutionary pathways via natural selection and / or artificial selection through domestication of the 5-HTT polymorphism, which might have enabled a stable social lifestyle in the wild ancestors of modern chickens.


Asunto(s)
Pollos/genética , Miedo/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Regiones no Traducidas 5'/genética , Alelos , Animales , Conducta Animal/fisiología , Miedo/psicología , Femenino , Genotipo , Polimorfismo Genético/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Conducta Social
19.
Curr Biol ; 28(19): 3136-3142.e4, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30245101

RESUMEN

Human and octopus lineages are separated by over 500 million years of evolution [1, 2] and show divergent anatomical patterns of brain organization [3, 4]. Despite these differences, growing evidence suggests that ancient neurotransmitter systems are shared across vertebrate and invertebrate species and in many cases enable overlapping functions [5]. Sociality is widespread across the animal kingdom, with numerous examples in both invertebrate (e.g., bees, ants, termites, and shrimps) and vertebrate (e.g., fishes, birds, rodents, and primates) lineages [6]. Serotonin is an evolutionarily ancient molecule [7] that has been implicated in regulating both invertebrate [8] and vertebrate [9] social behaviors, raising the possibility that this neurotransmitter's prosocial functions may be conserved across evolution. Members of the order Octopoda are predominantly asocial and solitary [10]. Although at this time it is unknown whether serotonergic signaling systems are functionally conserved in octopuses, ethological studies indicate that agonistic behaviors are suspended during mating [11-13], suggesting that neural mechanisms subserving social behaviors exist in octopuses but are suppressed outside the reproductive period. Here we provide evidence that, as in humans, the phenethylamine (+/-)-3,4-methylendioxymethamphetamine (MDMA) enhances acute prosocial behaviors in Octopus bimaculoides. This finding is paralleled by the evolutionary conservation of the serotonin transporter (SERT, encoded by the Slc6A4 gene) binding site of MDMA in the O. bimaculoides genome. Taken together, these data provide evidence that the neural mechanisms subserving social behaviors exist in O. bimaculoides and indicate that the role of serotonergic neurotransmission in regulating social behaviors is evolutionarily conserved.


Asunto(s)
Conducta Animal/fisiología , Neuronas Serotoninérgicas/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Conducta Agonística/efectos de los fármacos , Animales , Encéfalo/metabolismo , Evolución Molecular , N-Metil-3,4-metilenodioxianfetamina/farmacología , Neurotransmisores/metabolismo , Octopodiformes/fisiología , Filogenia , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Conducta Social , Transmisión Sináptica/fisiología
20.
Psychiatr Danub ; 30(2): 136-141, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29930222

RESUMEN

The microbiota-gut-brain axis is a bidirectional homeostatic route of communication between both of the organs direct via receptors of the CNS or via epigenetic mechanisms of divers metabolites e.g. SCFA, GABA, ß-hydroxybutyrate. Thus, a modulation of gut microbiota via nutrition, lifestyle etc. might be effective for emotional status and depressive disorders. The dietary composition has an influence on gut microbiota composition, microbial metabolite profile and the according consequences on emotional status and depression within a system biologic approach. There are changes in gut microbiota composition and gut microbial profile (butyrate, GABA, ß-hydroxybutyrate) effecting epigenetic regulation (histone acetylation, DNA methylation) and gene expression of receptors and mediators (SLC6A4, BDNF, GABA, GPRs) involved in depressive disorders.


Asunto(s)
Encéfalo/fisiopatología , Trastorno Depresivo/fisiopatología , Microbioma Gastrointestinal/fisiología , Neurotransmisores/fisiología , Ácido 3-Hidroxibutírico/fisiología , Acilación/fisiología , Butiratos/metabolismo , Metilación de ADN/fisiología , Trastorno Depresivo/genética , Emociones/fisiología , Epigénesis Genética/fisiología , Regulación de la Expresión Génica/fisiología , Histonas/fisiología , Homeostasis/fisiología , Humanos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/fisiología , Ácido gamma-Aminobutírico/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...