Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Int J Mol Sci ; 24(21)2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37958600

RESUMEN

Serotonin 1A (5-HT1A) autoreceptors located on serotonin neurons inhibit their activity, and their upregulation has been implicated in depression, suicide and resistance to antidepressant treatment. Conversely, post-synaptic 5-HT1A heteroreceptors are important for antidepressant response. The transcription factor deformed epidermal autoregulatory factor 1 (Deaf1) acts as a presynaptic repressor and postsynaptic enhancer of 5-HT1A transcription, but the mechanism is unclear. Because Deaf1 interacts with and is phosphorylated by glycogen synthase kinase 3ß (GSK3ß)-a constitutively active protein kinase that is inhibited by the mood stabilizer lithium at therapeutic concentrations-we investigated the role of GSK3ß in Deaf1 regulation of human 5-HT1A transcription. In 5-HT1A promoter-reporter assays, human HEK293 kidney and 5-HT1A-expressing SKN-SH neuroblastoma cells, transfection of Deaf1 reduced 5-HT1A promoter activity by ~45%. To identify potential GSK3ß site(s) on Deaf1, point mutations of known and predicted phosphorylation sites on Deaf1 were tested. Deaf1 repressor function was not affected by any of the mutants tested except the Y300F mutant, which augmented Deaf1 repression. Both lithium and the selective GSK3 inhibitors CHIR-99021 and AR-014418 attenuated and reversed Deaf1 repression compared to vector. This inhibition was at concentrations that maximally inhibit GSK3ß activity as detected by the GSK3ß-sensitive TCF/LEF reporter construct. Our results support the hypothesis that GSK3ß regulates the activity of Deaf1 to repress 5-HT1A transcription and provide a potential mechanism for actions of GSK3 inhibitors on behavior.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta , Litio , Receptor de Serotonina 5-HT1A , Serotonina , Humanos , Antidepresivos , Glucógeno Sintasa Quinasa 3 beta/genética , Células HEK293 , Litio/farmacología , Receptor de Serotonina 5-HT1A/genética , Serotonina/farmacología
2.
J Neurosci ; 38(38): 8200-8210, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30093565

RESUMEN

The serotonin-1A (5-HT1A) receptor is a key regulator of serotonergic activity and is implicated in mood and emotion. However, its post-transcriptional regulation has never been studied in humans. In the present study, we show that the "intronless" human 5-HT1A gene (HTR1A) is alternatively spliced in its 3'-UTR, yielding two novel splice variants. These variants lack a ∼1.6 kb intron, which contains an microRNA-135 (miR135) target site. Unlike the human HTR1A, the mouse HTR1A lacks the splice donor/accepter sites. Thus, in the mouse HTR1A, splicing was not detected. The two spliced mRNAs are extremely stable, are resistant to miR135-induced downregulation, and have greater translational output than the unspliced variant. Moreover, alternative HTR1A RNA splicing is oppositely regulated by the splice factors PTBP1 and nSR100, which inhibit or enhance its splicing, respectively. In postmortem human brain tissue from both sexes, HTR1A mRNA splicing was prevalent and region-specific. Unspliced HTR1A was expressed more strongly in the hippocampus and midbrain versus the prefrontal cortex (PFC), and correlated with reduced levels of nSR100. Importantly, HTR1A RNA splicing and nSR100 levels were reduced in the PFC of individuals with major depression compared with controls. Our unexpected findings uncover a novel mechanism to regulate HTR1A gene expression through alternative splicing of microRNA sites. Altered levels of splice factors could contribute to changes in regional and depression-related gene expression through alternative splicing.SIGNIFICANCE STATEMENT Alternative splicing, which is prevalent in brain tissue, increases gene diversity. The serotonin-1A receptor gene (HTR1A) is a regulator of serotonin, which is implicated in mood and emotion. Here we show that human HTR1A RNA is alternately spliced. Splicing removes a microRNA site to generate ultrastable RNA and increase HTR1A expression. This splicing varies in different brain regions and is reduced in major depression. We also identify specific splice factors for HTR1A RNA, showing they are also reduced in depression. Thus, we describe a novel mechanism to regulate gene expression through splicing. Altered levels of splice factors could contribute to depression by changing gene expression.


Asunto(s)
Empalme Alternativo , Trastorno Depresivo Mayor/metabolismo , Hipocampo/metabolismo , Mesencéfalo/metabolismo , Estabilidad del ARN/fisiología , Receptor de Serotonina 5-HT1A/metabolismo , Adulto , Trastorno Depresivo Mayor/genética , Femenino , Células HEK293 , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Receptor de Serotonina 5-HT1A/genética
3.
J Psychiatry Neurosci ; 44(3): 164-176, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30807072

RESUMEN

Major depression and anxiety are highly prevalent and involve chronic dysregulation of serotonin, but they remain poorly understood. Here, we review novel transcriptional (genetic, epigenetic) and posttranscriptional (microRNA, alternative splicing) mechanisms implicated in mental illness, focusing on a key serotonin-related regulator, the serotonin 1A (5-HT1A) receptor. Functional single-nucleotide polymorphisms and stress-induced DNA methylation of the 5-HT1A promoter converge to differentially alter pre- and postsynaptic 5-HT1A receptor expression associated with major depression and reduced therapeutic response to serotonergic antidepressants. Major depression is also associated with altered levels of splice factors and microRNA, posttranscriptional mechanisms that regulate RNA stability. The human 5-HT1A 3'-untranslated region is alternatively spliced, removing microRNA sites and increasing 5-HT1A expression, which is reduced in major depression and may be genotype-dependent. Thus, the 5-HT1A receptor gene illustrates the convergence of genetic, epigenetic and posttranscriptional mechanisms in gene expression, neurodevelopment and neuroplasticity, and major depression. Understanding gene regulatory mechanisms could enhance the detection, categorization and personalized treatment of major depression.


Asunto(s)
Empalme Alternativo/genética , Metilación de ADN/genética , Trastorno Depresivo Mayor/genética , Epigénesis Genética/genética , MicroARNs/genética , Receptor de Serotonina 5-HT1A/genética , Transcripción Genética/genética , Animales , Trastorno Depresivo Mayor/tratamiento farmacológico , Humanos
4.
Neurobiol Dis ; 82: 332-341, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26188176

RESUMEN

The serotonin 1A receptor (5-HT1A), a critical regulator of the brain serotonergic tone, is implicated in major depressive disorder (MDD) where it is often found to be dys-regulated. However, the extent to which stress and antidepressant treatment impact 5-HT1A expression in adults remains unclear. To address this issue, we subjected adult male BALB/c mice to unpredictable chronic mild stress (UCMS) to induce a depression-like phenotype that was reversed by chronic treatment with the antidepressant imipramine. In prefrontal cortex (PFC) and midbrain tissue, UCMS increased 5-HT1A RNA and protein levels, changes that are expected to decrease the brain serotonergic activity. The stress-induced increase in 5-HT1A expression was paralleled by a specific increase in DNA methylation of the conserved -681 CpG promoter site, located within a Sp1-like element. We show that the -681 CpG site is recognized and repressed by Sp4, the predominant neuronal Sp1-like factor and that Sp4-induced repression is attenuated by DNA methylation, despite a stress-induced increase in PFC Sp4 levels. These results indicate that adult life stress induces DNA methylation of a conserved promoter site, antagonizing Sp4 repression to increase 5-HT1A expression. Chronic imipramine treatment fully reversed the UCMS-induced increase in methylation of the -681 CpG site in the PFC but not midbrain of stressed animals and also increased 5-HT1A expression in the PFC of control animals. Incomplete reversal by imipramine of stress-induced changes in 5-HT1A methylation and expression indicates a persistence of stress vulnerability, and that sustained reversal of behavioral impairments may require additional pathways.


Asunto(s)
Antidepresivos Tricíclicos/farmacología , Metilación de ADN/efectos de los fármacos , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Receptor de Serotonina 5-HT1A/genética , Receptor de Serotonina 5-HT1A/metabolismo , Animales , Enfermedad Crónica , Secuencia Conservada , Islas de CpG , Metilación de ADN/fisiología , Trastorno Depresivo/genética , Modelos Animales de Enfermedad , Núcleo Dorsal del Rafe/efectos de los fármacos , Núcleo Dorsal del Rafe/metabolismo , Imipramina/farmacología , Masculino , Ratones Endogámicos BALB C , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología
5.
J Biol Chem ; 288(34): 24569-80, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23846693

RESUMEN

Double-stranded (ds) RNA of viral origin, a ligand for Melanoma Differentiation-associated gene 5 (MDA5) and Toll-Like Receptor 3 (TLR3), induces the TANK-Binding Kinase 1 (TBK1)-dependent phosphorylation and activation of Interferon Regulatory Factor 3 (IRF3) and the E3 ubiquitin ligase Pellino1, which are required for interferon ß (IFNß) gene transcription. Here, we report that Pellino1 interacts with the transcription factor Deformed Epidermal Autoregulatory Factor 1 (DEAF1). The interaction is independent of the E3 ligase activity of Pellino1, but weakened by the phosphorylation of Pellino1. We show that DEAF1 binds to the IFNß promoter and to IRF3 and IRF7, that it is required for the transcription of the IFNß gene and IFNß secretion in MEFs infected with Sendai virus or transfected with poly(I:C). DEAF1 is also needed for TLR3-dependent IFNß production. Taken together, our results identify DEAF1 as a novel component of the signal transduction network by which dsRNA of viral origin stimulates IFNß production.


Asunto(s)
Interferón beta/biosíntesis , Proteínas Nucleares/metabolismo , ARN Bicatenario/farmacología , Infecciones por Respirovirus/metabolismo , Virus Sendai/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Proteínas de Unión al ADN , Células HEK293 , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Interferón beta/genética , Ratones , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Infecciones por Respirovirus/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Ubiquitina-Proteína Ligasas/genética
6.
J Biol Chem ; 287(9): 6615-27, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22232550

RESUMEN

Altered regulation of the serotonin-1A (5-HT1A) receptor gene is implicated in major depression and mood disorders. The functional human 5-HT1A C(-1019)G promoter polymorphism (rs6295), which prevents the binding of Deaf-1/NUDR leading to dysregulation of the receptor, has been associated with major depression. In cell models Deaf-1 displays dual activity, repressing 5-HT1A autoreceptor expression in serotonergic raphe cells while enhancing postsynaptic 5-HT1A heteroreceptor expression in nonserotonergic neurons. A functional Deaf-1 binding site on the mouse 5-HT1A promoter was recognized by Deaf-1 in vitro and in vivo and mediated dual activity of Deaf-1 on 5-HT1A gene transcription. To address regulation by Deaf-1 in vivo, Deaf-1 knock-out mice bred to a C57BL/6 background were compared with wild-type siblings for changes in 5-HT1A RNA and protein by quantitative RT-PCR, in situ hybridization, and immunofluorescence. In the dorsal raphe, Deaf-1 knock-out mice displayed increased 5-HT1A mRNA, protein, and 5-HT1A-positive cell counts but reduced 5-HT levels, whereas other serotonergic markers, such as tryptophan hydroxylase (TPH)- or 5-HT-positive cells and TPH2 RNA levels, were unchanged. By contrast, 5-HT1A mRNA and 5-HT1A-positive cells were reduced in the frontal cortex of Deaf-1-null mice, with no significant change in hippocampal 5-HT1A RNA, protein, or cell counts. The region-specific alterations of brain 5-HT1A gene expression and reduced raphe 5-HT content in Deaf-1(-/-) mice indicate the importance of Deaf-1 in regulation of 5-HT1A gene expression and provide insight into the role of the 5-HT1A G(-1019) allele in reducing serotonergic neurotransmission by derepression of 5-HT1A autoreceptors.


Asunto(s)
Autorreceptores/genética , Núcleos del Rafe/fisiología , Receptor de Serotonina 5-HT1A/genética , Serotonina/metabolismo , Factores de Transcripción/genética , Animales , Autorreceptores/metabolismo , Proteínas de Unión al ADN , Trastorno Depresivo/metabolismo , Trastorno Depresivo/fisiopatología , Femenino , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo Genético/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Factores de Transcripción/metabolismo , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
7.
Microorganisms ; 11(5)2023 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-37317196

RESUMEN

Skin acts as a barrier that promotes the colonization of bacteria, fungi, archaea, and viruses whose membership and function may differ depending on the various specialized niches or micro-environments of the skin. The group of microorganisms inhabiting the skin, also known as the skin microbiome, offers protection against pathogens while actively interacting with the host's immune system. Some members of the skin microbiome can also act as opportunistic pathogens. The skin microbiome is influenced by factors such as skin site, birth mode, genetics, environment, skin products, and skin conditions. The association(s) of the skin microbiome with health and disease has (have) been identified and characterized via culture-dependent and culture-independent methods. Culture-independent methods (such as high-throughput sequencing), in particular, have expanded our understanding of the skin microbiome's role in maintaining health or promoting disease. However, the intrinsic challenges associated with the low microbial biomass and high host content of skin microbiome samples have hindered advancements in the field. In addition, the limitations of current collection and extraction methods and biases derived from sample preparation and analysis have significantly influenced the results and conclusions of many skin microbiome studies. Therefore, the present review discusses the technical challenges associated with the collection and processing of skin microbiome samples, the advantages and disadvantages of current sequencing approaches, and potential future areas of focus for the field.

8.
J Neurochem ; 116(6): 1066-76, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21182526

RESUMEN

Serotonin (5-hydroxytryptamine, 5-HT) neurotransmission is negatively regulated by 5-HT1A autoreceptors on raphe neurons, and is implicated in mood disorders. Pet-1/FEV is an ETS transcription factor expressed exclusively in serotonergic neurons and is essential for serotonergic differentiation, although its regulation of 5-HT receptors has not yet been studied. Here, we show by electrophoretic mobility shift assay that recombinant human Pet-1/FEV binds directly to multiple Pet-1 elements of the human 5-HT1A receptor promoter to enhance its transcriptional activity. In luciferase reporter assays, mutational analysis indicated that while several sites contribute, the Pet-1 site at -1406 bp had the greatest effect on 5-HT1A promoter activity. To address the effect of Pet-1 on 5-HT1A receptor regulation in vivo, we compared the expression of 5-HT1A receptor RNA and protein in Pet-1 null and wild-type littermate mice. In the raphe nuclei of Pet-1-/- mice tryptophan hydroxylase 2 (TPH2) RNA, and 5-HT and TPH immunostaining were greatly reduced, indicating a deficit in 5-HT production. Raphe 5-HT1A RNA and protein levels were also reduced in Pet-1-deficient mice, consistent with an absence of Pet-1-mediated transcriptional enhancement of 5-HT1A autoreceptors in serotonergic neurons. Interestingly, 5-HT1A receptor expression was up-regulated in the hippocampus, but down-regulated in the striatum and cortex. These data indicate that, in addition to transcriptional regulation by Pet-1 in raphe neurons, 5-HT1A receptor expression is regulated indirectly by alterations in 5-HT neurotransmission in a region-specific manner that together may contribute to the aggressive/anxiety phenotype observed in Pet-1 null mice.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica/fisiología , Receptor de Serotonina 5-HT1A/metabolismo , Factores de Transcripción/fisiología , Análisis de Varianza , Animales , Encéfalo/anatomía & histología , Línea Celular Transformada , Línea Celular Tumoral , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Noqueados , Neuroblastoma , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Ratas , Receptor de Serotonina 5-HT1A/genética , Factores de Transcripción/deficiencia , Transfección/métodos , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo
10.
Neuropharmacology ; 55(6): 977-85, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18639564

RESUMEN

The serotonin-1A (5-HT1A) receptor serves as a hub to regulate the activity and actions of the serotonin system, and is expressed both as a presynaptic autoreceptor on raphe neurons, and as a major postsynaptic receptor in hippocampal, cortical, and hypothalamic regions involved in mood, emotion and stress response. As such, the level of expression of 5-HT1A receptors is implicated in the development of anxiety and depression phenotypes. This review focuses on the C(-1019)G (rs6295) promoter polymorphism of the 5-HT1A receptor gene (HTR1A) and its effect on the activity of transcription factors that recognize the C-allele, including Deaf-1, Hes1 and Hes5; its effects on 5-HT1A receptor expression in pre- and postsynaptic areas; as well as its implication in early postnatal development and adult neurogenesis in the hippocampus and cortex. Although several studies have now replicated the association of the G-allele with depression, panic disorder, neuroticism, and reduced response to antidepressant or antipsychotic treatment, ethnic, disease and genetic heterogeneity among subjects in different studies may obscure such associations. Gene-gene interaction studies suggest that the 5-HT1A receptor G(-1019) allele is a risk allele which could be used as a marker for depression and related mood disorders. Finally, association of the G(-1019) allele with increased raphe 5-HT1A binding potential, increased amygdala reactivity to emotional stimuli, and reduced amygdala volume, particularly in disease states, suggests a functional role for the C(-1019)G site in 5-HT1A receptor dys-regulation and predisposition to mental illness.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Trastornos Mentales/genética , Polimorfismo Genético/genética , Receptor de Serotonina 5-HT1A/genética , Animales , Antidepresivos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Trastornos Mentales/patología
11.
Anticancer Res ; 28(4B): 2175-80, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18751392

RESUMEN

BACKGROUND: 5-Fluorouracil (5-FU) is an antineoplastic drug that targets thymidylate synthase (TS). Tumour cells can develop resistance to anti-TS drugs by a variety of mechanisms including up-regulation of TS protein and alterations in drug uptake and degradation. The possible mechanisms of the observed rapid development of resistance to the pyrimidine analogs 5-FUdR and 5-FU in cultured HCT116 colon cancer cells were investigated. MATERIALS AND METHODS: Cell survival was determined in resistant and control HCT116 cells treated with 5-FUdR and 5-FU for 7 days. The ability of the cells to take up and metabolize these drugs was determined by Western blotting and [3H]thymidine incorporation. RESULTS AND CONCLUSION: Resistant HCT116 cells were 5- and 100-fold more resistant to killing by 5-FU and 5-FUdR, respectively, than the parental cells and exhibited impaired uptake. Although the HCT116R cells were initially Mycoplasma free, a low level of Mycoplasma contamination was found in these cells after several weeks in culture. Sensitivity to 5-FUdR was restored by treatment with an anti-Mycoplasma antibiotic. Our observations emphasize the need for frequent testing for Mycoplasma contamination in any cell line under investigation for resistance to anti-TS drugs.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/microbiología , Floxuridina/farmacología , Fluorouracilo/farmacología , Infecciones por Mycoplasma/metabolismo , Aminopterina/metabolismo , Aminopterina/farmacología , Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos , Células HCT116 , Células HT29 , Células HeLa , Humanos , Hipoxantina/metabolismo , Hipoxantina/farmacología , Infecciones por Mycoplasma/tratamiento farmacológico , Timidina/metabolismo , Timidina/farmacología , Timidina Quinasa/metabolismo , Timidilato Sintasa/antagonistas & inhibidores , Timidilato Sintasa/metabolismo , Tritio
12.
Sci Rep ; 8(1): 5788, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636529

RESUMEN

The 5-HT1A autoreceptor mediates feedback inhibition of serotonin (5-HT) neurons, and is implicated in major depression. The human 5-HT1A gene (HTR1A) rs6295 risk allele prevents Deaf1 binding to HTR1A, resulting in increased 5-HT1A autoreceptor transcription. Since chronic stress alters HTR1A methylation and expression, we addressed whether recruitment of methyl-binding protein MeCP2 may alter Deaf1 regulation at the HTR1A locus. We show that MeCP2 enhances Deaf1 binding to its HTR1A site and co-immunoprecipitates with Deaf1 in cells and brain tissue. Chromatin immunoprecipitation assays showed Deaf1-dependent recruitment of MeCP2 to the mouse HTR1A promoter, and MeCP2 modulated human and mouse HTR1A gene transcription in a Deaf1-dependent fashion, enhancing Deaf1-induced repression at the Deaf1 site. To address the role of MeCP2 in HTR1A regulation in vivo, mice with conditional knockout of MeCP2 in adult 5-HT neurons (MeCP2 cKO) were generated. These mice exhibited increased 5-HT1A autoreceptor levels and function, consistent with MeCP2 enhancement of Deaf1 repression in 5-HT neurons. Interestingly, female MeCP2-cKO mice displayed reduced anxiety, while males showed increased anxiety and reduced depression-like behaviors. These data uncover a novel role for MeCP2 in 5-HT neurons to repress HTR1A expression and drive adult anxiety- and depression-like behaviors in a sex-specific manner.


Asunto(s)
Ansiedad/metabolismo , Depresión/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Receptor de Serotonina 5-HT1A/genética , Neuronas Serotoninérgicas/metabolismo , Animales , Ansiedad/genética , Encéfalo/metabolismo , Línea Celular , Proteínas de Unión al ADN , Depresión/genética , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Factores Sexuales , Factores de Transcripción/metabolismo
13.
Mol Brain ; 9(1): 77, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27488351

RESUMEN

The C (-1019) G rs6295 promoter polymorphism of the serotonin-1A (5-HT1A) receptor gene is associated with major depression in several but not all studies, suggesting that compensatory mechanisms mediate resilience. The rs6295 risk allele prevents binding of the repressor Deaf1 increasing 5-HT1A receptor gene transcription, and the Deaf1-/- mouse model shows an increase in 5-HT1A autoreceptor expression. In this study, Deaf1-/- mice bred on a mixed C57BL6-BALB/c background were compared to wild-type littermates for 5-HT1A autoreceptor function and behavior in males and females. Despite a sustained increase in 5-HT1A autoreceptor binding levels, the amplitude of the 5-HT1A autoreceptor-mediated current in 5-HT neurons was unaltered in Deaf1-/- mice, suggesting compensatory changes in receptor function. Consistent with increased 5-HT1A autoreceptor function in vivo, hypothermia induced by the 5-HT1A agonist DPAT was augmented in early generation male but not female Deaf1-/- mice, but was reduced with succeeding generations. Loss of Deaf1 resulted in a mild anxiety phenotype that was sex-and test-dependent, with no change in depression-like behavior. Male Deaf1 knockout mice displayed anxiety-like behavior in the open field and light-dark tests, while female Deaf1-/- mice showed increased anxiety only in the elevated plus maze. These data show that altered 5-HT1A autoreceptor regulation in male Deaf1-/- mice can be compensated for by generational adaptation of receptor response that may help to normalize behavior. The sex dependence of Deaf1 function in mice is consistent with a greater role for 5-HT1A autoreceptors in sensitivity to depression in men.


Asunto(s)
Adaptación Fisiológica , Ansiedad/fisiopatología , Receptor de Serotonina 5-HT1A/metabolismo , Caracteres Sexuales , Factores de Transcripción/deficiencia , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Ansiedad/complicaciones , Ansiedad/metabolismo , Conducta Animal/efectos de los fármacos , Proteínas de Unión al ADN , Depresión/complicaciones , Depresión/metabolismo , Depresión/fisiopatología , Femenino , Hipotermia Inducida , Activación del Canal Iónico/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Serotonina/farmacología , Factores de Transcripción/metabolismo
14.
Mol Brain ; 4: 21, 2011 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-21619616

RESUMEN

The serotonin-1A (5-HT1A) receptor is among the most abundant and widely distributed 5-HT receptors in the brain, but is also expressed on serotonin neurons as an autoreceptor where it plays a critical role in regulating the activity of the entire serotonin system. Over-expression of the 5-HT1A autoreceptor has been implicated in reducing serotonergic neurotransmission, and is associated with major depression and suicide. Extensive characterization of the transcriptional regulation of the 5-HT1A gene (HTR1A) using cell culture systems has revealed a GC-rich "housekeeping" promoter that non-selectively drives its expression; this is flanked by a series of upstream repressor elements for REST, Freud-1/CC2D1A and Freud-2/CC2D1B factors that not only restrict its expression to neurons, but may also regulate the level of expression of 5-HT1A receptors in various subsets of neurons, including serotonergic neurons. A separate set of allele-specific factors, including Deaf1, Hes1 and Hes5 repress at the HTR1A C(-1019)G (rs6295) polymorphism in serotonergic neurons in culture, as well as in vivo. Pet1, an obligatory enhancer for serotonergic differentiation, has been identified as a potent activator of 5-HT1A autoreceptor expression. Taken together, these results highlight an integrated regulation of 5-HT1A autoreceptors that differs in several aspects from regulation of post-synaptic 5-HT1A receptors, and could be selectively targeted to enhance serotonergic neurotransmission.


Asunto(s)
Autorreceptores/genética , Regulación de la Expresión Génica , Trastornos Mentales/genética , Receptor de Serotonina 5-HT1A/genética , Transcripción Genética , Autorreceptores/metabolismo , Humanos , Receptor de Serotonina 5-HT1A/metabolismo , Serotonina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA