Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Aesthetic Plast Surg ; 44(1): 162-167, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792563

RESUMEN

BACKGROUND: Fat grafting, used for soft tissue augmentation during aesthetic or reconstructive plastic surgery, has disadvantages of low efficiency and unpredictable resorption rate. As an alternative, cell-assisted lipotransfer (CAL) is widely used because of its simplicity and low fat resorption rate. However, relevant studies on optimal CAL parameters are still lacking. Here, we aimed to identify the most effective ratio of fat to stromal vascular fraction (SVF) for CAL. METHODS: We designed two experimental paradigms. The first involved four groups of mice, each group injected with varying ratios of fat and SVF purified from different amounts of fat from a fixed amount of harvested fat. The second experiment involved four groups of mice, each injected with varying amounts of SVF mixed with a fixed amount of fat tissue. The amount of surviving fat in both experiments was compared 8 weeks after fat transplantation. RESULTS: In the first experiment, the group injected with only fat, without consuming any of the harvested fat for SVF purification, showed the greatest mean volume and weight. In the second experiment, groups with 1:1 or more ratio of fat to SVF showed greater volume and weight than the group without SVF. Notably, a ratio of 1:1 did not give significantly different results than higher ratios. CONCLUSIONS: Thus, when a limited amount of fat tissue is available, using all of it for grafting is the most effective. However, if an adequate amount is available, using a fat-to-SVF ratio of 1:1 is the most efficient. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.


Asunto(s)
Tejido Adiposo , Procedimientos de Cirugía Plástica , Tejido Adiposo/trasplante , Animales , Estética , Ratones
2.
Ann Lab Med ; 44(5): 426-436, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38529546

RESUMEN

Background: Metformin, a drug prescribed for patients with type 2 diabetes, has potential efficacy in enhancing antitumor immunity; however, the detailed underlying mechanisms remain to be elucidated. Therefore, we aimed to identify the inhibitory molecular mechanisms of metformin on programmed death ligand 1 (PD-L1) expression in cancer cells and programmed death 1 (PD-1) expression in immune cells. Methods: We employed a luciferase reporter assay, quantitative real-time PCR, immunoblotting analysis, immunoprecipitation and ubiquitylation assays, and a natural killer (NK) cell-mediated tumor cell cytotoxicity assay. A mouse xenograft tumor model was used to evaluate the effect of metformin on tumor growth, followed by flow-cytometric analysis using tumor-derived single-cell suspensions. Results: Metformin decreased AKT-mediated ß-catenin S552 phosphorylation and subsequent ß-catenin transactivation in an adenosine monophosphate-activated protein kinase (AMPK) activation-dependent manner, resulting in reduced CD274 (encoding PD-L1) transcription in cancer cells. Tumor-derived soluble factors enhanced PD-1 protein stability in NK and T cells via dissociation of PD-1 from ubiquitin E3 ligases and reducing PD-1 polyubiquitylation. Metformin inhibited the tumor-derived soluble factor-reduced binding of PD-1 to E3 ligases and PD-1 polyubiquitylation, resulting in PD-1 protein downregulation in an AMPK activation-dependent manner. These inhibitory effects of metformin on both PD-L1 and PD-1 expression ameliorated cancer-reduced cytotoxic activity of immune cells in vitro and decreased tumor immune evasion and growth in vivo. Conclusions: Metformin blocks both PD-L1 and PD-1 within the tumor microenvironment. This study provided a mechanistic insight into the efficacy of metformin in improving immunotherapy in human cancer.


Asunto(s)
Antígeno B7-H1 , Células Asesinas Naturales , Metformina , Receptor de Muerte Celular Programada 1 , beta Catenina , Metformina/farmacología , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Humanos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Animales , Línea Celular Tumoral , Ratones , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , beta Catenina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas Activadas por AMP/metabolismo , Ratones Endogámicos BALB C , Fosforilación/efectos de los fármacos , Ratones Desnudos , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias/patología , Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Ubiquitinación/efectos de los fármacos
3.
Exp Mol Med ; 56(6): 1373-1387, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38825643

RESUMEN

N6-adenosine methylation (m6A) is critical for controlling cancer cell growth and tumorigenesis. However, the function and detailed mechanism of how m6A methyltransferases modulate m6A levels on specific targets remain unknown. In the current study, we identified significantly elevated levels of RBM15, an m6A writer, in basal-like breast cancer (BC) patients compared to nonbasal-like BC patients and linked this increase to worse clinical outcomes. Gene expression profiling revealed correlations between RBM15 and serine and glycine metabolic genes, including PHGDH, PSAT1, PSPH, and SHMT2. RBM15 influences m6A levels and, specifically, the m6A levels of serine and glycine metabolic genes via direct binding to target RNA. The effects of RBM15 on cell growth were largely dependent on serine and glycine metabolism. Thus, RBM15 coordinates cancer cell growth through altered serine and glycine metabolism, suggesting that RBM15 is a new therapeutic target in BC.


Asunto(s)
Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glicina , Proteínas de Unión al ARN , Serina , Neoplasias de la Mama Triple Negativas , Humanos , Serina/metabolismo , Glicina/metabolismo , Femenino , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Línea Celular Tumoral , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Metilación , Adenosina/metabolismo , Adenosina/análogos & derivados
4.
J Hazard Mater ; 464: 132966, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-37976851

RESUMEN

Exposure to ambient ultrafine particulate matter (UPM) causes respiratory disorders; however, the underlying molecular mechanisms remain unclear. In this study, we synthesized simulated UPM (sUPM) with controlled physicochemical properties using the spark-discharge method. Subsequently, we investigated the biological effects of sUPM using BEAS-2B human bronchial epithelial cells (HBECs) and a mouse intratracheal instillation model. High throughput RNA-sequencing and bioinformatics analyses revealed that dysregulation of the glycolytic metabolism is involved in the inhibited proliferation and survival of HBECs by sUPM treatment. Furthermore, signaling pathway and enzymatic analyses showed that the treatment of BEAS-2B cells with sUPM induces the inactivation of extracellular signal-regulated kinase (ERK) and protein kinase B (PKB, also known as AKT), resulting in the downregulation of phosphofructokinase 2 (PFK2) S483 phosphorylation, PFK enzyme activity, and aerobic glycolysis in HBECs in an oxidative stress-independent manner. Additionally, intratracheal instillation of sUPM reduced the phosphorylation of ERK, AKT, and PFK2, decreased proliferation, and increased the apoptosis of bronchial epithelial cells in mice. The findings of this study imply that UPM induces pulmonary toxicity by disrupting aerobic glycolytic metabolism in lung epithelial cells, which can provide novel insights into the toxicity mechanisms of UPM and strategies to prevent their toxic effects.


Asunto(s)
Contaminantes Atmosféricos , Material Particulado , Humanos , Animales , Ratones , Material Particulado/análisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosforilación , Células Epiteliales , Glucólisis , Fosfofructoquinasas/análisis , Fosfofructoquinasas/metabolismo , Contaminantes Atmosféricos/análisis
5.
ACS Macro Lett ; 12(5): 659-666, 2023 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-37155320

RESUMEN

Electrospinning has shown great potential for the fabrication of 3D nanofibrous tubular scaffolds for bifurcated vascular grafts. However, fabrication of complex 3D nanofibrous tubular scaffolds with bifurcated or patient-specific shapes remains limited. In this study, a 3D hollow nanofibrous bifurcated-tubular scaffold was fabricated by the uniform and conformal deposition of electrospun nanofibers via conformal electrospinning. By conformal electrospinning, electrospun nanofibers are conformally deposited onto a complex shape, such as the bifurcated region, without large pores or defects. Owing to conformal electrospinning, a corner profile fidelity (FC), a measure of conformal deposition of electrospun nanofibers at the bifurcated region, was increased 4 times at the bifurcation angle (θB) of 60°, and all FC values of the scaffolds reached 100%, regardless of the θB. Furthermore, the thickness of the scaffolds could be controlled by varying the electrospinning time. Leakage-free liquid transfer was successfully achieved owing to the uniform and conformal deposition of electrospun nanofibers. Finally, the cytocompatibility and 3D mesh-based modeling of the scaffolds were demonstrated. Thus, conformal electrospinning can be used to fabricate leakage-free and complex 3D nanofibrous scaffolds for bifurcated vascular grafts.

6.
J Exp Clin Cancer Res ; 42(1): 340, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38098117

RESUMEN

BACKGROUND: Cancer cells undergo cellular adaptation through metabolic reprogramming to sustain survival and rapid growth under various stress conditions. However, how brain tumors modulate their metabolic flexibility in the naturally serine/glycine (S/G)-deficient brain microenvironment remain unknown. METHODS: We used a range of primary/stem-like and established glioblastoma (GBM) cell models in vitro and in vivo. To identify the regulatory mechanisms of S/G deprivation-induced metabolic flexibility, we employed high-throughput RNA-sequencing, transcriptomic analysis, metabolic flux analysis, metabolites analysis, chromatin immunoprecipitation (ChIP), luciferase reporter, nuclear fractionation, cycloheximide-chase, and glucose consumption. The clinical significances were analyzed in the genomic database (GSE4290) and in human GBM specimens. RESULTS: The high-throughput RNA-sequencing and transcriptomic analysis demonstrate that the de novo serine synthesis pathway (SSP) and glycolysis are highly activated in GBM cells under S/G deprivation conditions. Mechanistically, S/G deprivation rapidly induces reactive oxygen species (ROS)-mediated AMP-activated protein kinase (AMPK) activation and AMPK-dependent hypoxia-inducible factor (HIF)-1α stabilization and transactivation. Activated HIF-1α in turn promotes the expression of SSP enzymes phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase 1 (PSAT1), and phosphoserine phosphatase (PSPH). In addition, the HIF-1α-induced expression of glycolytic genes (GLUT1, GLUT3, HK2, and PFKFB2) promotes glucose uptake, glycolysis, and glycolytic flux to fuel SSP, leading to elevated de novo serine and glycine biosynthesis, NADPH/NADP+ ratio, and the proliferation and survival of GBM cells. Analyses of human GBM specimens reveal that the levels of overexpressed PHGDH, PSAT1, and PSPH are positively correlated with levels of AMPK T172 phosphorylation and HIF-1α expression and the poor prognosis of GBM patients. CONCLUSION: Our findings reveal that metabolic stress-enhanced glucose-derived de novo serine biosynthesis is a critical metabolic feature of GBM cells, and highlight the potential to target SSP for treating human GBM.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Glioblastoma , Humanos , Glioblastoma/patología , Serina , Glucosa/metabolismo , Glicina , ARN , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Línea Celular Tumoral , Microambiente Tumoral , Fosfofructoquinasa-2
7.
Exp Cell Res ; 314(10): 2055-65, 2008 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-18445495

RESUMEN

The leucine-rich repeat kinase 2 (LRRK2) has been identified as the defective gene at the PARK8 locus causing the autosomal dominant form of Parkinson's disease (PD). Although several LRRK2 mutations were found in familial as well as sporadic PD patients, its physiological functions are not clearly defined. In this study, using yeast two-hybrid screening, we report the identification of Rab5b as an LRRK2-interacting protein. Indeed, our GST pull down and co-immunoprecipitation assays showed that it specifically interacts with LRRK2. In addition, subcellular fractionation and immunocytochemical analyses confirmed that a fraction of both proteins co-localize in synaptic vesicles. Interestingly, we found that alteration of LRRK2 expression by either overexpression or knockdown of endogenous LRRK2 in primary neuronal cells significantly impairs synaptic vesicle endocytosis. Furthermore, this endocytosis defect was rescued by co-expression of functional Rab5b protein, but not by its inactive form. Taken together, we propose that LRRK2, in conjunction with its interaction with Rab5b, plays an important role in synaptic function by modulating the endocytosis of synaptic vesicles.


Asunto(s)
Endocitosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Vesículas Sinápticas/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Animales , Línea Celular , Exocitosis/fisiología , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Neuronas/citología , Neuronas/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ratas , Sinapsis/metabolismo , Técnicas del Sistema de Dos Híbridos , Proteínas de Unión al GTP rab5/genética
8.
Artículo en Inglés | MEDLINE | ID: mdl-31438592

RESUMEN

Expected climatic changes likely elicit serious challenges for crop production. Therefore, it is indispensable to investigate the response of crop growth parameters and yield under temperature variability environments. The current experiment on chilli pepper growth was conducted in a field, rain-shelter plastic house, and plastic greenhouse, with accumulated temperatures of 2832 °C, 2967 °C, and 3105 °C in 2017; and 2944 °C, 3091 °C, and 3168 °C in 2018 growing seasons. Based on soil analysis, 132.7 kg ha-1 (1× of livestock manure compost as an optimum and 265.4 kg ha-1 (2×) as a double amount of organic matter were applied to each simulated temperature condition. The results showed that organic manure application favorably affects the growth attributes and nutrient uptake of chilli pepper with the highest values found in the plastic greenhouse, followed by the rain-shelter house, over the open field cultivation condition. The highest growth of chilli pepper was at the 2× rate of organic manure application, whereas the highest yield was found at the 1× rate of organic manure application. The application of organic manure at the 1× rate in the greenhouse increased root, shoot, and fruit dry weights of chilli pepper by 21.4%, 52.4%, and 79.7%, respectively, compared to the control values. These results indicate that the rational use of organic amendments might be the best solution for chilli pepper production under variable climate conditions.


Asunto(s)
Capsicum/crecimiento & desarrollo , Fertilizantes , Estiércol , Frutas/crecimiento & desarrollo , Raíces de Plantas/crecimiento & desarrollo , Brotes de la Planta/crecimiento & desarrollo , Estaciones del Año , Suelo/química , Temperatura
9.
Exp Mol Med ; 40(3): 345-53, 2008 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-18587273

RESUMEN

For cancer gene therapy, cancer-specific over- expression of a therapeutic gene is required to reduce side effects derived from expression of the gene in normal cells. To develop such an expression vector, we searched for genes over-expressed and/or specifically expressed in cancer cells using bioinformatics and have selected genes coding for protein regulator of cytokinesis 1 (PRC1) and ribonuclease reductase 2 (RRM2) as candidates. Their cancer-specific expressions were confirmed in both breast cancer cell lines and patient tissues. We compared each promoter's cancer-specific activity in the breast normal and cancer cell lines using the luciferase gene as a reporter and confirmed cancer-specific expression of both PRC1 and RRM2 promoters. To test activities of these promoters in viral vectors, the promoters were also cloned into an adeno-associated viral (AAV) vector containing green fluorescence protein (GFP) as the reporter. The GFP expression levels by these promoters were various depending on cell lines tested and, in MDA-MB-231 cells, GFP activities derived from the PRC1 and RRM2 promoters were as strong as that from the cytomegalovirus (CMV) promoter. Our result showed that a vector containing the PRC1 or RRM2 promoter could be used for breast cancer specific overexpression in gene therapy.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Ciclo Celular/genética , Marcación de Gen , Regiones Promotoras Genéticas/genética , Ribonucleósido Difosfato Reductasa/genética , Activación Transcripcional , Neoplasias de la Mama/terapia , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Clonación Molecular , Citomegalovirus , Dependovirus , Femenino , Terapia Genética , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Ribonucleósido Difosfato Reductasa/metabolismo
10.
J Biochem ; 157(6): 485-95, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25605758

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) has been identified as a causative gene for Parkinson's disease (PD). LRRK2 contains a kinase and a GTPase domain, both of which provide critical intracellular signal-transduction functions. We showed previously that Rab5b, a small GTPase protein that regulates the motility and fusion of early endosomes, interacts with LRRK2 and co-regulates synaptic vesicle endocytosis. Using recombinant proteins, we show here that LRRK2 phosphorylates Rab5b at its Thr6 residue in in vitro kinase assays with mass spectrophotometry analysis. Phosphorylation of Rab5b by LRRK2 on the threonine residue was confirmed by western analysis using cells stably expressing LRRK2 G2019S. The phosphomimetic T6D mutant exhibited stronger GTPase activity than that of the wild-type Rab5b. In addition, phosphorylation of Rab5b by LRRK2 also exhibited GTPase activity stronger than that of the unphosphorylated Rab5b protein. Two assays testing Rab5's activity, neurite outgrowth analysis and epidermal growth factor receptor degradation assays, showed that Rab5b T6D exhibited phenotypes that were expected to be observed in the inactive Rab5b, including longer neurite length and less degradation of EGFR. These results suggest that LRRK2 kinase activity functions as a Rab5b GTPase activating protein and thus, negatively regulates Rab5b signalling.


Asunto(s)
Endosomas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Receptores ErbB/metabolismo , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Imitación Molecular , Fosforilación , Especificidad por Sustrato
11.
Exp Mol Med ; 45: e36, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23949442

RESUMEN

Leucine-rich repeat kinase 2 (LRRK2) is a gene that, upon mutation, causes autosomal-dominant familial Parkinson's disease (PD). Yeast two-hybrid screening revealed that Snapin, a SNAP-25 (synaptosomal-associated protein-25) interacting protein, interacts with LRRK2. An in vitro kinase assay exhibited that Snapin is phosphorylated by LRRK2. A glutathione-S-transferase (GST) pull-down assay showed that LRRK2 may interact with Snapin via its Ras-of-complex (ROC) and N-terminal domains, with no significant difference on interaction of Snapin with LRRK2 wild type (WT) or its pathogenic mutants. Further analysis by mutation study revealed that Threonine 117 of Snapin is one of the sites phosphorylated by LRRK2. Furthermore, a Snapin T117D phosphomimetic mutant decreased its interaction with SNAP-25 in the GST pull-down assay. SNAP-25 is a component of the SNARE (Soluble NSF Attachment protein REceptor) complex and is critical for the exocytosis of synaptic vesicles. Incubation of rat brain lysate with recombinant Snapin T117D, but not WT, protein caused decreased interaction of synaptotagmin with the SNARE complex based on a co-immunoprecipitation assay. We further found that LRRK2-dependent phosphorylation of Snapin in the hippocampal neurons resulted in a decrease in the number of readily releasable vesicles and the extent of exocytotic release. Combined, these data suggest that LRRK2 may regulate neurotransmitter release via control of Snapin function by inhibitory phosphorylation.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Secuencia de Aminoácidos , Animales , Exocitosis , Femenino , Células HEK293 , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Fosforilación , Fosfotreonina/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Qa-SNARE/metabolismo , Ratas , Ratas Sprague-Dawley , Sinaptotagminas/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , Proteínas de Transporte Vesicular/química
12.
BMB Rep ; 46(9): 454-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24064060

RESUMEN

LRRK2 (leucine-rich repeat kinase 2) has been identified as a gene corresponding to PARK8, an autosomal-dominant gene for familial Parkinson's disease (PD). LRRK2 pathogenic-specific mutants induce neurotoxicity and shorten neurites. To elucidate the mechanism underlying LRRK2 expression, we constructed the LRRK2-promoter-luciferase reporter and used it for promoter analysis. We found that the glucocorticoid receptor (GR) transactivated LRRK2 in a ligand-dependent manner. Using quantitative RT-PCR and Western analysis, we further showed that treatment with dexamethasone, a synthetic GR ligand, induced LRRK2 expression at both the transcriptional and translational levels, in dopaminergic MN9D cells. Dexamethasone treatment also increased expression of α α-synuclein, another PD causative gene, and enhanced transactivation of the α-synuclein promoter-luciferase reporter. In addition, dexamethasone treatment to MN9D cells weakly induced cytotoxicity based on an LDH assay. Because glucocorticoid hormones are secreted in response to stress, our data suggest that stress might be a related factor in the pathogenesis of PD.


Asunto(s)
Dexametasona/farmacología , Expresión Génica/efectos de los fármacos , Glucocorticoides/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Dexametasona/toxicidad , Glucocorticoides/toxicidad , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Ratones , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Receptores de Glucocorticoides/metabolismo , Activación Transcripcional/efectos de los fármacos , alfa-Sinucleína/genética
13.
Biol Chem ; 389(10): 1313-8, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18713018

RESUMEN

The constitutive androstane receptor (CAR) is a member of the nuclear receptor superfamily and plays an important role in the degradation of xenobiotics in the liver. Using yeast two-hybrid screening, we identified SF3a3, a 60-kDa subunit of the splicing factor 3a complex, as a specific CAR-interacting protein. We further confirmed their interaction by both co-immunoprecipitation and GST pull-down assay. Functional studies showed that overexpression of SF3a3 inhibited the reporter activity driven by a promoter containing CAR binding sequences by up to 50%, whereas reduced expression of SF3a3 activated the same reporter activity by approximately three-fold. The inhibitory function of SF3a3 is independent of the presence of TCPOBOP, a CAR ligand. These data suggest that SF3a3 functions as a co-repressor of CAR transcriptional activity, in addition to its canonical function.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Receptor de Androstano Constitutivo , Vectores Genéticos , Humanos , Ratones , Receptores Citoplasmáticos y Nucleares/genética , Ribonucleoproteína Nuclear Pequeña U2/genética , Factores de Transcripción/genética , Transcripción Genética , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA