Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(1)2023 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-38203414

RESUMEN

The HSP70 and HSP90 family members belong to molecular chaperones that exhibit protective functions during the cellular response to stressful agents. We investigated whether the exposure of rats to chronic mild stress (CMS), a validated model of depression, affects the expression of HSP70 and HSP90 in the prefrontal cortex (PFC), hippocampus (HIP) and thalamus (Thal). Male Wistar rats were exposed to CMS for 3 or 8 weeks. The antidepressant imipramine (IMI, 10 mg/kg, i.p., daily) was introduced in the last five weeks of the long-term CMS procedure. Depressive-like behavior was verified by the sucrose consumption test. The expression of mRNA and protein was quantified by real-time PCR and Western blot, respectively. In the 8-week CMS model, stress alone elevated HSP72 and HSP90B mRNA expression in the HIP. HSP72 mRNA was increased in the PFC and HIP of rats not responding to IMI treatment vs. IMI responders. The CMS exposure increased HSP72 protein expression in the cytosolic fraction of the PFC and HIP, and this effect was diminished by IMI treatment. Our results suggest that elevated levels of HSP72 may serve as an important indicator of neuronal stress reactions accompanying depression pathology and could be a potential target for antidepressant strategy.


Asunto(s)
Imipramina , Chaperonas Moleculares , Masculino , Ratas , Animales , Imipramina/farmacología , Ratas Wistar , Proteínas HSP70 de Choque Térmico , Hipocampo , Proteínas HSP90 de Choque Térmico/genética , Corteza Prefrontal , ARN Mensajero/genética , Antidepresivos/farmacología
2.
Neurochem Int ; 155: 105302, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35150790

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor deficits caused by the loss of dopaminergic neurons in the substantia nigra (SN) and ventral tegmental area (VTA). However, clinical data revealed that not only the dopaminergic system is affected in PD. Postmortem studies showed degeneration of noradrenergic cells in the locus coeruleus (LC) to an even greater extent than that observed in the SN/VTA. Pharmacological models support the concept that modification of noradrenergic transmission can influence the PD-like phenotype induced by neurotoxins. Nevertheless, there are no existing data on animal models regarding the distant impact of noradrenergic degeneration on intact SN/VTA neurons. The aim of this study was to create a transgenic mouse model with endogenously evoked progressive degeneration restricted to noradrenergic neurons and investigate its long-term impact on the dopaminergic system. To this end, we selectively ablated the transcription initiation factor-IA (TIF-IA) in neurons expressing dopamine ß-hydroxylase (DBH) by the Cre-loxP system. This mutation mimics a condition of nucleolar stress affecting neuronal survival. TIF-IADbhCre mice were characterized by selective, progressive degeneration of noradrenergic neurons, followed by phenotypic alterations associated with sympathetic system impairment. Our studies did not show any loss of tyrosine hydroxylase (TH)-positive cells in the SN/VTA of mutant mice; however, we observed increased indices of oxidative stress, enhanced markers of glial cell activation, inflammatory processes and isolated degenerating cells positive for FluoroJade C. These results were supported by gene expression profiling of VTA and SN from TIF-IADbhCre mice, revealing that 34 out of 246 significantly regulated genes in the SN/VTA were related to PD. Overall, our results shed new light on the possible negative influence of noradrenergic degeneration on dopaminergic neurons, reinforcing the neuroprotective role of noradrenaline.


Asunto(s)
Mesencéfalo , Sustancia Negra , Animales , Neuronas Dopaminérgicas/metabolismo , Inflamación/metabolismo , Ratones , Norepinefrina/metabolismo , Estrés Oxidativo , Sustancia Negra/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Área Tegmental Ventral/metabolismo
3.
Int J Mol Sci ; 22(9)2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062902

RESUMEN

Currently utilized antidepressants have limited effectiveness and frequently incur undesired effects. Most antidepressants are thought to act via the inhibition of monoamine reuptake; however, direct binding to monoaminergic receptors has been proposed to contribute to both their clinical effectiveness and their side effects, or lack thereof. Among the target receptors of antidepressants, α1­adrenergic receptors (ARs) have been implicated in depression etiology, antidepressant action, and side effects. However, differences in the direct effects of antidepressants on signaling from the three subtypes of α1-ARs, namely, α1A-, α1B- and α1D­ARs, have been little explored. We utilized cell lines overexpressing α1A-, α1B- or α1D-ARs to investigate the effects of the antidepressants imipramine (IMI), desipramine (DMI), mianserin (MIA), reboxetine (REB), citalopram (CIT) and fluoxetine (FLU) on noradrenaline-induced second messenger generation by those receptors. We found similar orders of inhibition at α1A-AR (IMI < DMI < CIT < MIA < REB) and α1D­AR (IMI = DMI < CIT < MIA), while the α1B-AR subtype was the least engaged subtype and was inhibited with low potency by three drugs (MIA < IMI = DMI). In contrast to their direct antagonistic effects, prolonged incubation with IMI and DMI increased the maximal response of the α1B-AR subtype, and the CIT of both the α1A- and the α1B-ARs. Our data demonstrate a complex, subtype-specific modulation of α1-ARs by antidepressants of different groups.


Asunto(s)
Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Receptores Adrenérgicos alfa 1/genética , Animales , Antidepresivos/clasificación , Citalopram/farmacología , Depresión/etiología , Depresión/genética , Depresión/patología , Desipramina/farmacología , Fluoxetina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Imipramina/farmacología , Mianserina/farmacología , Ratones , Células PC12 , Ratas , Reboxetina/farmacología , Transducción de Señal/efectos de los fármacos
4.
Pharmacol Rep ; 73(4): 1179-1187, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34117630

RESUMEN

BACKGROUND: Evidence indicates that Gα12, Gα13, and its downstream effectors, RhoA and Rac1, regulate neuronal morphology affected by stress. This study was aimed at investigating whether repeated stress influences the expression of proteins related to the Gα12/13 intracellular signaling pathway in selected brain regions sensitive to the effects of stress. Furthermore, the therapeutic impact of ß(1)adrenergic receptors (ß1AR) blockade was assessed. METHODS: Restraint stress (RS) model in mice (2 h/14 days) was used to assess prolonged stress effects on the mRNA expression of Gα12, Gα13, RhoA, Rac1 in the prefrontal cortex (PFC), hippocampus (HIP) and amygdala (AMY). In a separate study, applying RS model in rats (3-4 h/1 day or 14 days), we evaluated stress effects on the expression of Gα12, Gα11, Gαq, RhoA, RhoB, RhoC, Rac1/2/3 in the HIP. Betaxolol (BET), a selective ß1AR antagonist, was introduced (5 mg/kg/p.o./8-14 days) in the rat RS model to assess the role of ß1AR in stress effects. RT-qPCR and Western Blot were used for mRNA and protein assessments, respectively. RESULTS: Chronic RS decreased mRNA expression of Gα12 and increased mRNA for Rac1 in the PFC of mice. In the mice AMY, decreased mRNA expression of Gα12, Gα13 and RhoA was observed. Fourteen days of RS exposure increased RhoA protein level in the rats' HIP in the manner dependent on ß1AR activity. CONCLUSIONS: Together, these results suggest that repeated RS affects the expression of genes and proteins known to be engaged in neural plasticity, providing potential targets for further studies aimed at unraveling the molecular mechanisms of stress-related neuropsychiatric diseases.


Asunto(s)
Encéfalo/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Neuronas/metabolismo , Restricción Física/fisiología , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Antagonistas de Receptores Adrenérgicos beta 1/farmacología , Animales , Encéfalo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores Adrenérgicos beta 1/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Pharmacol Rep ; 71(5): 753-761, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31351316

RESUMEN

BACKGROUND: The transcription factor CREB and the neurotrophin BDNF are important mood regulators due to their profound role in controlling the neuronal plasticity. Our previously published results from transgenic mice functionally lacking CREB in chosen neural populations have shown that BDNF upregulation evoked by chronic treatment with fluoxetine seems to be dependent on CREB residing exclusively in serotonergic neurons. To further elucidate this observation, we focused on the representative signaling cascades engaged in the regulation of BDNF production. METHODS: The study was carried out on mice lacking CREB in noradrenergic (Creb1DBHCre) or serotonergic (Creb1TPH2CreERT2) neurons in CREM deficient background. Animals received fluoxetine (10 mg/kg, ip) or desipramine (20 mg/kg, ip) for 21 days. The expression of following proteins and their phosphorylated forms was assessed by Western blot: CREB, BDNF, CaMKIIα, ERK1/2. RESULTS: We showed that consistent with previously observed BDNF upregulation, chronic treatment with fluoxetine causes an increase in the pool of active CaMKIIα in w/t males, while in Creb1TPH2CreERT2 mutants, this effect ceased along with the observed decrease in ERK1/2 phosphorylation. These effects were region- and sex-specific. We did not observe a similar pattern of changes regarding the levels of BDNF expression and the CaMKIIα, ERK1/2 kinases in Creb1DBHCre mice exposed to desipramine. However, sex-dependent changes in the regulation of CaMKIIα and ERK1/2 activity were also observed. CONCLUSIONS: Our study highlights the pivotal role of CREB in response to antidepressants, emphasizing different sex-dependent vulnerabilities to particular drugs and the important impact of CREM on the effects of CREB deletion.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neuronas Serotoninérgicas/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Animales , Antidepresivos/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Desipramina/farmacología , Femenino , Fluoxetina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Neuronas Serotoninérgicas/efectos de los fármacos , Caracteres Sexuales
6.
Artículo en Inglés | MEDLINE | ID: mdl-30953677

RESUMEN

Disturbances in fear-evoked signal transduction in the hippocampus (HP), the nuclei of the amygdala (AMY), and the prefrontal cortex (PFC) underlie anxiety-related disorders. However, the molecular mechanisms underlying these effects remain elusive. Heterotrimeric G proteins (GPs) are divided into the following four families based on the intracellular activity of their alpha subunit (Gα): Gα(s) proteins stimulate cyclic AMP (cAMP) generation, Gα(i/o) proteins inhibit the cAMP pathway, Gα(q/11) proteins increase the intracellular Ca++ concentration and the inositol trisphosphate level, and Gα(12/13) proteins activate monomeric GP-Rho. In the present study, we assessed the effects of a fear memory procedure on the mRNA expression of the Gα subunits of all four GP families in the HP, AMY and PFC. C57BL/6 J mice were subjected to a fear conditioning (FC) procedure followed by a contextual or cued fear memory test (CTX-R and CS-R, respectively). Morphine (MOR, 1 mg/kg/ip) was injected immediately after FC to prevent the fear consolidation process. Real-time quantitative PCR was used to measure the mRNA expression levels of Gα subunits at 1 h after FC, 24 h after FC, and 1 h after the CTX-R or CS-R. In the HP, the mRNA levels of Gα(s), Gα(12) and Gα(11) were higher at 1 h after training. Gα(s) levels were slightly lower when consolidation was stabilized and after the CS-R. The mRNA levels of Gα(12) were increased at 1 h after FC, returned to control levels at 24 h after FC and increased again with the CTX-R. The increase in the Gα(11) level persisted at 24 h after FC and after CTX-R. In the AMY, no specific changes were induced by FC. In the PFC, CTX-R was accompanied by a decrease in Gα(i/o) mRNA levels; however, only Gα(i2) downregulation was prevented by MOR treatment. Hence, the FC-evoked changes in Gα mRNA expression were observed mainly in the HP and connected primarily to contextual learning. These results suggest that the activation of signaling pathways by Gα(s) and Gα(12) is required to begin the fear memory consolidation process in the HP, while signal transduction via Gα(11) is implicated in the maintenance of fear consolidation. In the PFC, the downregulation of Gα(i2) appears to be related to the contextual learning of fear.


Asunto(s)
Encéfalo/metabolismo , Miedo , Proteínas de Unión al GTP/metabolismo , Memoria/efectos de los fármacos , Morfina/farmacología , Animales , Encéfalo/efectos de los fármacos , Condicionamiento Clásico/efectos de los fármacos , Señales (Psicología) , Miedo/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo
7.
Sci Rep ; 9(1): 5262, 2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30918302

RESUMEN

Parkinson's disease (PD) is the second most common neurodegenerative disorder and is characterized by motor deficits such as tremor, rigidity and bradykinesia. These symptoms are directly caused by the loss of dopaminergic neurons. However, a wealth of clinical evidence indicates that the dopaminergic system is not the only system affected in PD. Postmortem studies of brains from PD patients have revealed the degeneration of noradrenergic neurons in the locus coeruleus (LC) to the same or even greater extent than that observed in the dopaminergic neurons of substantia nigra (SN) and ventral tegmental area (VTA). Moreover, studies performed on rodent models suggest that enhancement of noradrenergic transmission may attenuate the PD-like phenotype induced by MPTP administration, a neurotoxin-based PD model. The aim of this study was to investigate whether chronic treatment with either of two compounds targeting the noradrenergic system (reboxetine or atipamezole) possess the ability to reduce the progression of a PD-like phenotype in a novel mouse model of progressive dopaminergic neurodegeneration induced by the genetic inhibition of rRNA synthesis in dopaminergic neurons, mimicking a PD-like phenotype. The results showed that reboxetine improved the parkinsonian phenotype associated with delayed progression of SN/VTA dopaminergic neurodegeneration and higher dopamine content in the striatum. Moreover, the alpha1-adrenergic agonist phenylephrine enhanced survival of TH+ neurons in primary cell cultures, supporting the putative neuroprotective effects of noradrenergic stimulation. Our results provide new insights regarding the possible influence of the noradrenergic system on dopaminergic neuron survival and strongly support the hypothesis regarding the neuroprotective role of noradrenaline.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/metabolismo , Reboxetina/uso terapéutico , Animales , Células Cultivadas , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Femenino , Imidazoles/uso terapéutico , Inmunohistoquímica , Locus Coeruleus/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Sustancia Negra/efectos de los fármacos , Sustancia Negra/metabolismo , Área Tegmental Ventral/citología
8.
Front Neurosci ; 12: 637, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30294251

RESUMEN

Neurotrophic factors are regarded as crucial regulatory components in neuronal plasticity and are postulated to play an important role in depression pathology. The abundant expression of brain-derived neurotrophic factor (BDNF) in various brain structures seems to be of particular interest in this context, as downregulation of BDNF is postulated to be correlated with depression and its upregulation is often observed after chronic treatment with common antidepressants. It is well-known that BDNF expression is regulated by cyclic AMP response element-binding protein (CREB). In our previous study using mice lacking CREB in serotonergic neurons (Creb1TPH2CreERT2 mice), we showed that selective CREB ablation in these particular neuronal populations is crucial for drug-resistant phenotypes in the tail suspension test observed after fluoxetine administration in Creb1TPH2CreERT2 mice. The aim of this study was to investigate the molecular changes in the expression of neurotrophins in Creb1TPH2CreERT2 mice after chronic fluoxetine treatment, restricted to the brain structures implicated in depression pathology with profound serotonergic innervation including the prefrontal cortex (PFC) and hippocampus. Here, we show for the first time that BDNF upregulation observed after fluoxetine in the hippocampus or PFC might be dependent on the transcription factor CREB residing, not within these particular structures targeted by serotonergic projections, but exclusively in serotonergic neurons. This observation may shed new light on the neurotrophic hypothesis of depression, where the effects of BDNF observed after antidepressants in the hippocampus and other brain structures were rather thought to be regulated by CREB residing within the same brain structures. Overall, these results provide further evidence for the pivotal role of CREB in serotonergic neurons in maintaining mechanisms of antidepressant drug action by regulation of BDNF levels.

9.
Sci Rep ; 7(1): 13515, 2017 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-29044198

RESUMEN

Evidence exists that chronic antidepressant therapy enhances CREB levels and activity. Nevertheless, the data are not conclusive, as previous analysis of transgenic mouse models has suggested that CREB inactivation in fact contributes to antidepressant-like behavior. The aim of this study was to evaluate the role of CREB in this context by exploiting novel transgenic mouse models, characterized by selective ablation of CREB restricted to noradrenergic (Creb1DBHCre/Crem-/-) or serotonergic (Creb1TPH2CreERT2/Crem-/-) neurons in a CREM-deficient background to avoid possible compensatory effects of CREM. Selective and functional ablation of CREB affected antidepressant-like behavior in a tail suspension test (TST) after antidepressant treatment. Contrary to single Creb1DBHCre mutants, Creb1DBHCre/Crem-/- mice did not respond to acute desipramine administration (20 mg/kg) on the TST. On the other hand, single Creb1TPH2CreERT2 mutants displayed reduced responses to fluoxetine (10 mg/kg) on the TST, while the effects in Creb1TPH2CreERT2/Crem-/- mice differed by gender. Our results provide further evidence for the important role of CREM as a compensatory factor. Additionally, the results indicate that new models based on the functional ablation of CREB in select neuronal populations may represent a valuable tool for investigating the role of CREB in the mechanism of antidepressant therapy.


Asunto(s)
Neuronas Adrenérgicas/metabolismo , Antidepresivos/uso terapéutico , Modulador del Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Depresión/genética , Neuronas Serotoninérgicas/metabolismo , Animales , Modulador del Elemento de Respuesta al AMP Cíclico/deficiencia , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/deficiencia , Depresión/tratamiento farmacológico , Depresión/etiología , Desipramina/uso terapéutico , Femenino , Fluoxetina/uso terapéutico , Suspensión Trasera/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL
10.
PeerJ ; 5: e3240, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28462043

RESUMEN

BACKGROUND: Huntington's disease (HD) is a rare familial autosomal dominant neurodegenerative disorder characterized by progressive degeneration of medium spiny neurons (MSNs) located in the striatum. Currently available treatments of HD are only limited to alleviating symptoms; therefore, high expectations for an effective therapy are associated with potential replacement of lost neurons through stimulation of postnatal neurogenesis. One of the drugs of potential interest for the treatment of HD is riluzole, which may act as a positive modulator of adult neurogenesis, promoting replacement of damaged MSNs. The aim of this study was to evaluate the effects of chronic riluzole treatment on a novel HD-like transgenic mouse model, based on the genetic ablation of the transcription factor TIF-IA. This model is characterized by selective and progressive degeneration of MSNs. METHODS: Selective ablation of TIF-IA in MSNs (TIF-IAD1RCre mice) was achieved by Cre-based recombination driven by the dopamine 1 receptor (D1R) promoter in the C57Bl/6N mouse strain. Riluzole was administered for 14 consecutive days (5 mg/kg, i.p.; 1× daily) starting at six weeks of age. Behavioral analysis included a motor coordination test performed on 13-week-old animals on an accelerated rotarod (4-40 r.p.m.; 5 min). To visualize the potential effects of riluzole treatment, the striata of the animals were stained by immunohistochemistry (IHC) and/or immunofluorescence (IF) with Ki67 (marker of proliferating cells), neuronal markers (NeuN, MAP2, DCX), and markers associated with neurodegeneration (GFAP, 8OHdG, FluoroJade C). Additionally, the morphology of dendritic spines of neurons was assessed by a commercially available FD Rapid Golgi Stain™ Kit. RESULTS: A comparative analysis of IHC staining patterns with chosen markers for the neurodegeneration process in MSNs did not show an effect of riluzole on delaying the progression of MSN cell death despite an observed enhancement of cell proliferation as visualized by the Ki67 marker. A lack of a riluzole effect was also reflected by the behavioral phenotype associated with MSN degeneration. Moreover, the analysis of dendritic spine morphology did not show differences between mutant and control animals. DISCUSSION: Despite the observed increase in newborn cells in the subventricular zone (SVZ) after riluzole administration, our study did not show any differences between riluzole-treated and non-treated mutants, revealing a similar extent of the neurodegenerative phenotype evaluated in 13-week-old TIF-IAD1RCre animals. This could be due to either the treatment paradigm (relatively low dose of riluzole used for this study) or the possibility that the effects were simply too weak to have any functional meaning. Nevertheless, this study is in line with others that question the effectiveness of riluzole in animal models and raise concerns about the utility of this drug due to its rather modest clinical efficacy.

11.
Eur Neuropsychopharmacol ; 24(9): 1546-56, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25106693

RESUMEN

It has been shown that stressful events occurring in early life have a powerful influence on the development of the central nervous system. Insulin-like growth factor-1 (IGF-1) promotes the growth, differentiation and survival of both neurons and glial cells and is thought to exert antidepressant-like activity. Thus, it is possible that disturbances in the function of the IGF-1 system may be responsible for disturbances observed over the course of depression. Prenatal stress was used as a valid model of depression. Adult male offspring of control and stressed rat dams were subjected to behavioural testing (forced swim test). The level of IGF-1 in the blood and the expression of IGF-1, IGF-1R, and IRS-1/2 in the hippocampus and frontal cortex using RT-PCR, ELISA and western blotting were measured. In addition the effect of intracerebroventricularly administered IGF-1 and/or the IGF-1R receptor antagonist JB1 in the forced swim test was studied. Prenatally stressed rats showed depressive like behaviour, including increased immobility time as well as decreased mobility and climbing. Intracerebroventricular administration of IGF-1 reversed these effects in stressed animals, whereas concomitant administration of the IGF-1R antagonist JB1 completely blocked the effects. Biochemical analysis of homogenates from the hippocampus and frontal cortex revealed decreases in IGF-1 level and IGF-1R phosphorylation along with disturbances in IRS-1 phosphorylation. These findings reveal that prenatal stress alters IGF-1 signalling, which may contribute to the behavioural changes observed in depression.


Asunto(s)
Encéfalo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Receptor IGF Tipo 1/metabolismo , Estrés Psicológico/etiología , Estrés Psicológico/patología , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Femenino , Inyecciones Intraventriculares , Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Péptidos/farmacología , Fosforilación/efectos de los fármacos , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Natación/psicología , Factores de Tiempo
12.
Pharmacol Rep ; 65(6): 1489-97, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24552996

RESUMEN

α1-Adrenergic receptors (α1-ARs) are important players in peripheral and central nervous system (CNS) regulation and function and in mediating various behavioral responses. The α1-AR family consists of three subtypes, α1A, α1B and α1D, which differ in their subcellular distribution, efficacy in evoking intracellular signals and transcriptional profiles. All three α1-AR subtypes are present at relatively high densities throughout the CNS, but the contributions of the individual subtypes to various central functions are currently unclear. Because of the lack of specific ligands, functionally characterizing the α1-ARs and discriminating between the three subtypes are difficult. To date, studies using genetically engineered mice have provided some information on subtype-related functions of the CNS α1-ARs. In this mini-review, we discuss several CNS processes where the α1-ARs role has been delineated with pharmacological tools and by studies using mutated mice strains that infer specific α1-AR subtype functions through evaluation of behavioral phenotypes.


Asunto(s)
Adrenérgicos/uso terapéutico , Animales Modificados Genéticamente/metabolismo , Sistema Nervioso Central/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Adrenérgicos/farmacocinética , Animales , Sistema Nervioso Central/efectos de los fármacos , Humanos , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...