Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neurochem Res ; 46(7): 1830-1843, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33881662

RESUMEN

Steroid receptor coactivator 1 (SRC-1) is one of the coactivators recruited by the nuclear receptors (NRs) when NRs are activated by steroid hormones, such as glucocorticoid. SRC-1 is abundant in hippocampus and hypothalamus and is also related to some major risk factors for depression, implicated by its reduced expression after stress and its effect on hypothalamus-pituitary-adrenal gland axis function. However, whether SRC-1 is involved in the formation of depression remains unclear. In this study, we firstly established chronic unpredictable stress (CUS) to induce depressive-like behaviors in mice and found that SRC-1 expression was reduced by CUS. A large number of studies have shown that neuroinflammation is associated with stress-induced depression and lipopolysaccharide (LPS) injection can lead to neuroinflammation and depressive-like behaviors in mice. Our result indicated that LPS treatment also decreased SRC-1 expression in mouse brain, implying the involvement of SRC-1 in the process of inflammation and depression. Next, we showed that the chronic unpredictable mild stress (CUMS) failed to elicit the depressive-like behaviors and dramatically promoted the expression of SRC-1 in brain of wild type mice. What's more, the SRC-1 knockout mice were more susceptible to CUMS to develop depressive-like behaviors and presented the changed expression of glucocorticoid receptor. However, SRC-1 deficiency did not affect the microglia activation induced by CUMS. Altogether, these results indicate a correlation between SRC-1 level and depressive-like behaviors, suggesting that SRC-1 might be involved in the development of depression induced by stress.


Asunto(s)
Depresión/metabolismo , Coactivador 1 de Receptor Nuclear/deficiencia , Estrés Psicológico/metabolismo , Animales , Células Cultivadas , Depresión/etiología , Femenino , Suspensión Trasera , Hipocampo/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/metabolismo , Coactivador 1 de Receptor Nuclear/metabolismo , Embarazo , Estrés Psicológico/complicaciones
2.
Neuroscience ; 377: 114-125, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29524638

RESUMEN

Steroids have been demonstrated to play profound roles in the regulation of hippocampal function by acting on their receptors, which need coactivators for their transcriptional activities. Previous studies have shown that steroid receptor coactivator-1 (SRC-1) is the predominant coactivator in the hippocampus, but its exact role and the underlying mechanisms remain unclear. In this study, we constructed SRC-1 RNA interference (RNAi) lentiviruses, injected them into the hippocampus of male mice, and then examined the changes in the expression of selected synaptic proteins, CA1 synapse density, postsynaptic density (PSD) thickness, and in vivo long-term potentiation (LTP). Spatial learning and memory behavior changes were investigated using the Morris water maze. We then transfected the lentiviruses into cultured hippocampal cells and examined the changes in synaptic protein and phospho-cyclic AMP response element-binding protein (pCREB) expression. The in vivo results showed that SRC-1 knockdown significantly decreased the expression of synaptic proteins and CA1 synapse density as well as PSD thickness; SRC-1 knockdown also significantly impaired in vivo LTP and disrupted spatial learning and memory. The in vitro results showed that while the expression of synaptic proteins was significantly decreased by SRC-1 knockdown, pCREB expression was also significantly decreased. The above results suggest a pivotal role of SRC-1 in the regulation of hippocampal synaptic plasticity and spatial learning and memory, strongly indicating SRC-1 may serve as a novel therapeutic target for hippocampus-dependent memory disorders.


Asunto(s)
Hipocampo/metabolismo , Potenciación a Largo Plazo/fisiología , Trastornos de la Memoria/metabolismo , Coactivador 1 de Receptor Nuclear/deficiencia , Memoria Espacial/fisiología , Animales , Línea Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Técnicas de Silenciamiento del Gen , Hipocampo/patología , Masculino , Trastornos de la Memoria/patología , Ratones Endogámicos C57BL , Coactivador 1 de Receptor Nuclear/genética , Fosforilación , Células Piramidales/metabolismo , Células Piramidales/patología , Interferencia de ARN , Aprendizaje Espacial/fisiología , Sinapsis/metabolismo , Sinapsis/patología
3.
J Clin Invest ; 125(7): 2808-24, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26098214

RESUMEN

The precise mechanisms that lead to parturition are incompletely defined. Surfactant protein-A (SP-A), which is secreted by fetal lungs into amniotic fluid (AF) near term, likely provides a signal for parturition; however, SP-A-deficient mice have only a relatively modest delay (~12 hours) in parturition, suggesting additional factors. Here, we evaluated the contribution of steroid receptor coactivators 1 and 2 (SRC-1 and SRC-2), which upregulate SP-A transcription, to the parturition process. As mice lacking both SRC-1 and SRC-2 die at birth due to respiratory distress, we crossed double-heterozygous males and females. Parturition was severely delayed (~38 hours) in heterozygous dams harboring SRC-1/-2-deficient embryos. These mothers exhibited decreased myometrial NF-κB activation, PGF2α, and expression of contraction-associated genes; impaired luteolysis; and elevated circulating progesterone. These manifestations also occurred in WT females bearing SRC-1/-2 double-deficient embryos, indicating that a fetal-specific defect delayed labor. SP-A, as well as the enzyme lysophosphatidylcholine acyltransferase-1 (LPCAT1), required for synthesis of surfactant dipalmitoylphosphatidylcholine, and the proinflammatory glycerophospholipid platelet-activating factor (PAF) were markedly reduced in SRC-1/-2-deficient fetal lungs near term. Injection of PAF or SP-A into AF at 17.5 days post coitum enhanced uterine NF-κB activation and contractile gene expression, promoted luteolysis, and rescued delayed parturition in SRC-1/-2-deficient embryo-bearing dams. These findings reveal that fetal lungs produce signals to initiate labor when mature and that SRC-1/-2-dependent production of SP-A and PAF is crucial for this process.


Asunto(s)
Intercambio Materno-Fetal/fisiología , Coactivador 1 de Receptor Nuclear/fisiología , Coactivador 2 del Receptor Nuclear/fisiología , Parto/fisiología , 1-Acilglicerofosfocolina O-Aciltransferasa/deficiencia , 1-Acilglicerofosfocolina O-Aciltransferasa/genética , Animales , Femenino , Madurez de los Órganos Fetales , Heterocigoto , Pulmón/embriología , Pulmón/fisiología , Luteólisis , Masculino , Intercambio Materno-Fetal/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Coactivador 1 de Receptor Nuclear/deficiencia , Coactivador 1 de Receptor Nuclear/genética , Coactivador 2 del Receptor Nuclear/deficiencia , Coactivador 2 del Receptor Nuclear/genética , Factor de Activación Plaquetaria/deficiencia , Embarazo , Regiones Promotoras Genéticas , Proteína A Asociada a Surfactante Pulmonar/deficiencia , Transducción de Señal , Activación Transcripcional , Útero/fisiología
4.
Mol Endocrinol ; 27(2): 366-80, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23315938

RESUMEN

The rapidly growing family of transcriptional coregulators includes coactivators that promote transcription and corepressors that harbor the opposing function. In recent years, coregulators have emerged as important regulators of metabolic homeostasis, including the p160 steroid receptor coactivator (SRC) family. Members of the SRC family have been ascribed important roles in control of gluconeogenesis, fat absorption and storage in the liver, and fatty acid oxidation in skeletal muscle. To provide a deeper and more granular understanding of the metabolic impact of the SRC family members, we performed targeted metabolomic analyses of key metabolic byproducts of glucose, fatty acid, and amino acid metabolism in mice with global knockouts (KOs) of SRC-1, SRC-2, or SRC-3. We measured amino acids, acyl carnitines, and organic acids in five tissues with key metabolic functions (liver, heart, skeletal muscle, brain, plasma) isolated from SRC-1, -2, or -3 KO mice and their wild-type littermates under fed and fasted conditions, thereby unveiling unique metabolic functions of each SRC. Specifically, SRC-1 ablation revealed the most significant impact on hepatic metabolism, whereas SRC-2 appeared to impact cardiac metabolism. Conversely, ablation of SRC-3 primarily affected brain and skeletal muscle metabolism. Surprisingly, we identified very few metabolites that changed universally across the three SRC KO models. The findings of this Research Resource demonstrate that coactivator function has very limited metabolic redundancy even within the homologous SRC family. Furthermore, this work also demonstrates the use of metabolomics as a means for identifying novel metabolic regulatory functions of transcriptional coregulators.


Asunto(s)
Metaboloma , Coactivadores de Receptor Nuclear/metabolismo , Aminoácidos/metabolismo , Animales , Encéfalo/metabolismo , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Coactivador 1 de Receptor Nuclear/deficiencia , Coactivador 1 de Receptor Nuclear/genética , Coactivador 2 del Receptor Nuclear/deficiencia , Coactivador 2 del Receptor Nuclear/genética , Coactivador 3 de Receptor Nuclear/deficiencia , Coactivador 3 de Receptor Nuclear/genética , Coactivadores de Receptor Nuclear/sangre , Coactivadores de Receptor Nuclear/genética , Plasma/metabolismo
5.
J Hepatol ; 56(1): 210-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21827731

RESUMEN

BACKGROUND & AIMS: Nuclear receptors such as pregnane X receptor and constitutive androstane receptor (CAR) are important regulators of drug-metabolizing systems such as P450 enzymes and modulate xenobiotic metabolism as well as hepatocellular proliferation. Binding of CAR to NR response elements alone is not sufficient to activate gene expression. Here, we investigate the role of steroid receptor co-activator (SRC) family members in CAR-mediated hepatocyte proliferation and drug metabolism. METHODS: The role of SRCs in CAR activation was assessed in cell-based transfection assays and protein-protein interaction assays. The in vivo role of SRCs in CAR-mediated hepatocyte proliferation and drug metabolism was examined by using mice deficient in SRCs. RESULTS: SRC-3 displayed the highest co-activating activity to CAR compared with SRC-1 and SRC-2 in a cell-based reporter assay. Knockout of SRC-3 in mice attenuated hepatic hyperplasia induced by a CAR agonist 1,4-bis-[2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP), which was associated with a reduced expression of c-Myc and Foxm-1. In contrast, knockout of SRC-1 or SRC-2 in mice did not affect TCPOBOP-induced hepatic hyperplasia. SRC-3-deficient mice were hypersensitive to zoxazolamine-induced paralysis, but were resistant to acetaminophen hepatotoxicity induced by TCPOBOP, whereas mutant mice deficient in SRC-1 or SRC-2 exhibited severe acetaminophen hepatotoxicity similar to wild-type controls. Accordingly, deficiency in SRC-3, but not SRC-1 or SRC-2, resulted in a reduced CAR-mediated expression of drug metabolism-related genes in the liver. CONCLUSIONS: Our study demonstrates that SRC-3 is the predominant transcriptional co-activator among the three SRC family members for CAR activation to promote hepatocyte proliferation and drug metabolism.


Asunto(s)
Hepatocitos/citología , Hepatocitos/metabolismo , Coactivador 3 de Receptor Nuclear/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Acetaminofén/toxicidad , Animales , Proliferación Celular , Receptor de Androstano Constitutivo , Expresión Génica , Hiperplasia , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Coactivador 1 de Receptor Nuclear/deficiencia , Coactivador 1 de Receptor Nuclear/genética , Coactivador 1 de Receptor Nuclear/metabolismo , Coactivador 2 del Receptor Nuclear/deficiencia , Coactivador 2 del Receptor Nuclear/genética , Coactivador 2 del Receptor Nuclear/metabolismo , Coactivador 3 de Receptor Nuclear/deficiencia , Coactivador 3 de Receptor Nuclear/genética , Piridinas/toxicidad , Xenobióticos/metabolismo
6.
BMC Cancer ; 10: 629, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21080969

RESUMEN

BACKGROUND: The peroxisome proliferator activated receptor (PPAR) subgroup of the nuclear hormone receptor superfamily is activated by a variety of natural and synthetic ligands. PPARs can heterodimerize with retinoid X receptors, which have homology to other members of the nuclear receptor superfamily. Ligand binding to PPAR/RXRs results in recruitment of transcriptional coactivator proteins such as steroid receptor coactivator 1 (SRC-1) and CREB binding protein (CBP). Both SRC-1 and CBP are histone acetyltransferases, which by modifying nucleosomal histones, produce more open chromatin structure and increase transcriptional activity. Nuclear hormone receptors can recruit limiting amounts of coactivators from other transcription factor binding sites such as AP-1, thereby inhibiting the activity of AP-1 target genes. PPAR and RXR ligands have been used in experimental breast cancer therapy. The role of coactivator expression in mammary tumorigenesis and response to drug therapy has been the subject of recent studies. METHODS: We examined the effects of loss of SRC-1 on MMTV-neu mediated mammary tumorigenesis. RESULTS: SRC-1 null mutation in mammary tumor prone mice increased the tumor latency period, reduced tumor proliferation index and metastasis, inhibited response to PPAR and RXR ligands, and induced genes involved in mammary gland differentiation. We also examined human breast cancer cell lines overexpressing SRC-1 or CBP. Coactivator overexpression increased cellular proliferation with resistance to PPAR and RXR ligands and remodeled chromatin of the proximal epidermal growth factor receptor promoter. CONCLUSIONS: These results indicate that histone acetyltransferases play key roles in mammary tumorigenesis and response to anti-proliferative therapies.


Asunto(s)
Antineoplásicos/farmacología , Genes erbB-2 , Neoplasias Mamarias Experimentales/prevención & control , Virus del Tumor Mamario del Ratón/genética , Coactivador 1 de Receptor Nuclear/deficiencia , Receptores Activados del Proliferador del Peroxisoma/agonistas , Animales , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Clofibrato/farmacología , Ácidos Grasos Insaturados/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Invasividad Neoplásica , Coactivador 1 de Receptor Nuclear/genética , Receptores Activados del Proliferador del Peroxisoma/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Regiones Promotoras Genéticas , Receptores X Retinoide/agonistas , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Tetrahidronaftalenos/farmacología , Tiazolidinedionas/farmacología , Factores de Tiempo , Transfección , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA