Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Craniofac Surg ; 29(6): 1629-1632, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29771839

RESUMEN

Distraction osteogenesis a surgical procedure conducted to improve craniofacial deformities. Compared with conventional operations, this technique has advantages such as the ability to lengthen the soft tissue and hard tissue. Therefore, this method is used to treat severe craniofacial abnormalities.The major disadvantage of distraction osteogenesis is the long treatment period. If the consolidation period is not sufficiently long after the distraction period, complications such as discontinuity or contraction of newly formed bone may occur. Recently, many researchers have attempted to develop methods for enhancing the ossification of newly formed bone, reducing shrinkage, and shortening the overall treatment period.The authors injected autologous bone marrow-derived mesenchymal stem cells during the consolidation phase after distraction osteogenesis surgery in a bilateral mandibular hypoplasia patient. Here, the authors report the treatment results, which were found to be favorable.


Asunto(s)
Cuidados Posteriores/métodos , Anomalías Craneofaciales/cirugía , Mandíbula , Trasplante de Células Madre Mesenquimatosas/métodos , Micrognatismo , Osteogénesis por Distracción/métodos , Adulto , Femenino , Humanos , Mandíbula/anomalías , Mandíbula/diagnóstico por imagen , Mandíbula/cirugía , Micrognatismo/diagnóstico , Micrognatismo/cirugía , Osteogénesis/fisiología , Resultado del Tratamiento
2.
Cancer Cell ; 10(6): 487-99, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17157789

RESUMEN

A serious obstacle to successful treatment of estrogen receptor (ER)-positive human breast cancer is cell resistance to tamoxifen (TAM) therapy. Here we show that the electrophile disulfide benzamide (DIBA), an ER zinc finger inhibitor, blocks ligand-dependent and -independent cell growth of TAM-resistant breast cancer in vitro and in vivo. Such inhibition depends on targeting disruption of the ER DNA-binding domain and its communication with neighboring functional domains, facilitating ERalpha dissociation from its coactivator AIB1 and concomitant association with its corepressor NCoR bound to chromatin. DIBA does not affect phosphorylation of HER2, MAPK, AKT, and AIB1, suggesting that DIBA-modified ERalpha may induce a switch from agonistic to antagonistic effects of TAM on resistant breast cancer cells.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Benzamidas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , ADN/metabolismo , Receptor alfa de Estrógeno/fisiología , Tamoxifeno/farmacología , Animales , Sitios de Unión , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Receptor alfa de Estrógeno/química , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Ratones , Fosforilación , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-2/metabolismo , Elementos de Respuesta
3.
J Cell Mol Med ; 16(2): 273-86, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21362131

RESUMEN

Vascular calcification is prevalent in patients with chronic kidney disease and leads to increased cardiovascular morbidity and mortality. Although several reports have implicated mitochondrial dysfunction in cardiovascular disease and chronic kidney disease, little is known about the potential role of mitochondrial dysfunction in the process of vascular calcification. This study investigated the effect of α-lipoic acid (ALA), a naturally occurring antioxidant that improves mitochondrial function, on vascular calcification in vitro and in vivo. Calcifying vascular smooth muscle cells (VSMCs) treated with inorganic phosphate (Pi) exhibited mitochondrial dysfunction, as demonstrated by decreased mitochondrial membrane potential and ATP production, the disruption of mitochondrial structural integrity and concurrently increased production of reactive oxygen species. These Pi-induced functional and structural mitochondrial defects were accompanied by mitochondria-dependent apoptotic events, including release of cytochrome c from the mitochondria into the cytosol, subsequent activation of caspase-9 and -3, and chromosomal DNA fragmentation. Intriguingly, ALA blocked the Pi-induced VSMC apoptosis and calcification by recovery of mitochondrial function and intracellular redox status. Moreover, ALA inhibited Pi-induced down-regulation of cell survival signals through the binding of growth arrest-specific gene 6 (Gas6) to its cognate receptor Axl and subsequent Akt activation, resulting in increased survival and decreased apoptosis. Finally, ALA significantly ameliorated vitamin D(3) -induced aortic calcification and mitochondrial damage in mice. Collectively, the findings suggest ALA attenuates vascular calcification by inhibiting VSMC apoptosis through two distinct mechanisms; preservation of mitochondrial function via its antioxidant potential and restoration of the Gas6/Axl/Akt survival pathway.


Asunto(s)
Mitocondrias/metabolismo , Músculo Liso Vascular/metabolismo , Ácido Tióctico/metabolismo , Calcificación Vascular/metabolismo , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células Cultivadas , Colecalciferol/farmacología , Citocromos c , Fragmentación del ADN , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/enzimología , Mitocondrias/patología , Músculo Liso Vascular/patología , Fosfatos/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo , Tirosina Quinasa del Receptor Axl
4.
Circ Res ; 104(7): 842-50, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19229058

RESUMEN

Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are important pathogenic mechanisms in atherosclerosis and restenosis after vascular injury. In this study, we investigated the effects of beta-lapachone (betaL) (3,4-Dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran-5,6-dione), which is a potent antitumor agent that stimulates NAD(P)H:quinone oxidoreductase (NQO)1 activity, on neointimal formation in animals given vascular injury and on the proliferation of VSMCs cultured in vitro. betaL significantly reduced the neointimal formation induced by balloon injury. betaL also dose-dependently inhibited the FCS- or platelet-derived growth factor-induced proliferation of VSMCs by inhibiting G(1)/S phase transition. betaL increased the phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase 1 in rat and human VSMCs. Chemical inhibitors of AMPK or dominant-negative AMPK blocked the betaL-induced suppression of cell proliferation and the G(1) cell cycle arrest, in vitro and in vivo. The activation of AMPK in VSMCs by betaL is mediated by LKB1 in the presence of NQO1. Taken together, these results show that betaL inhibits VSMCs proliferation via the NQO1 and LKB1-dependent activation of AMPK. These observations provide the molecular basis that pharmacological stimulation of NQO1 activity is a new therapy for the treatment of vascular restenosis and/or atherosclerosis which are caused by proliferation of VSMCs.


Asunto(s)
Traumatismos de las Arterias Carótidas/tratamiento farmacológico , Estenosis Carotídea/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Activadores de Enzimas/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/farmacología , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Traumatismos de las Arterias Carótidas/enzimología , Traumatismos de las Arterias Carótidas/patología , Estenosis Carotídea/enzimología , Estenosis Carotídea/patología , Ciclo Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Activación Enzimática , Activadores de Enzimas/toxicidad , Inhibidores Enzimáticos/farmacología , Células HeLa , Humanos , Hiperplasia , Masculino , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NAD(P)H Deshidrogenasa (Quinona)/genética , Naftoquinonas/toxicidad , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína de Retinoblastoma/metabolismo , Prevención Secundaria , Factores de Tiempo , Proteína p53 Supresora de Tumor/metabolismo , Túnica Íntima/efectos de los fármacos , Túnica Íntima/enzimología , Túnica Íntima/patología
5.
Arterioscler Thromb Vasc Biol ; 30(11): 2164-72, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20829507

RESUMEN

OBJECTIVE: To explore whether α-lipoic acid (ALA), a naturally occurring antioxidant, inhibits neointimal hyperplasia by inducing apoptosis of vascular smooth muscle cells and to examine its potential effects on reendothelialization and platelet aggregation. METHODS AND RESULTS: Restenosis and late stent thrombosis, caused by neointimal hyperplasia and delayed reendothelialization, are significant clinical problems of balloon angioplasty and drug-eluting stents. ALA treatment strongly induced apoptosis of vascular smooth muscle cells and enhanced the expression and cytoplasmic localization of Nur77, which triggers intrinsic apoptotic events. Small interfering RNA-mediated downregulation of Nur77 diminished this proapoptotic effect of ALA. Moreover, ALA increased p38 mitogen-activated protein kinase phosphorylation, and inhibition of p38 mitogen-activated protein kinase completely blocked ALA-induced vascular smooth muscle cell apoptosis and Nur77 induction and cytoplasmic localization. In balloon-injured rat carotid arteries, ALA enhanced Nur77 expression and increased TUNEL-positive apoptotic cells in the neointima, leading to inhibition of neointimal hyperplasia. This preventive effect of ALA was significantly reduced by infection of an adenovirus encoding Nur77 small hairpin (sh)RNA. Furthermore, ALA reduced basal apoptosis of human aortic endothelial cells and accelerated reendothelialization after balloon injury. ALA also suppressed arachidonic acid-induced platelet aggregation. CONCLUSIONS: ALA could be a promising therapeutic agent to prevent restenosis and late stent thrombosis after angioplasty and drug-eluting stent implantation.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Ácido Tióctico/farmacología , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Células Endoteliales , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/lesiones , Hiperplasia/prevención & control , Masculino , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Ratas , Cicatrización de Heridas/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 29(10): 1558-64, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19696405

RESUMEN

OBJECTIVE: Clusterin is induced in vascular smooth muscle cells (VSMCs) during atherosclerosis and injury-induced neointimal hyperplasia. However, its functional roles in VSMCs and endothelial cells remain controversial and elusive. This study was undertaken to clarify the role of clusterin in neointimal hyperplasia and elucidate its mechanism of action. METHODS AND RESULTS: Adenovirus-mediated overexpression of clusterin (Ad-Clu) repressed TNF-alpha-stimulated expression of MCP-1, fractalkine, ICAM-1, VCAM-1, and MMP-9, leading to inhibition of VSMC migration. Both Ad-Clu and secreted clusterin suppressed VSMC proliferation by inhibiting DNA synthesis, but not by inducing apoptosis. Ad-Clu upregulated p53 and p21(cip1/waf1) but downregulated cyclins D and E, leading to suppression of pRb phosphorylation and subsequent induction of G1 arrest in VSMCs. Clusterin deficiency augmented VSMC proliferation in vitro and accelerated neointimal hyperplasia in vivo, but concomitantly impaired reendothelialization in wire-injured murine femoral arteries. Moreover, Ad-Clu significantly reduced neointimal thickening in balloon-injured rat carotid arteries. Clusterin also diminished TNF-alpha-induced apoptosis of human umbilical vein endothelial cells and restored endothelial nitric oxide synthase expression suppressed by TNF-alpha. CONCLUSIONS: These results suggest that upregulation of clusterin during vascular injury may be a protective response against, rather than a causative response to, the development of neointimal hyperplasia.


Asunto(s)
Clusterina/fisiología , Citoprotección , Células Endoteliales/citología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/fisiología , Túnica Íntima/patología , Animales , Movimiento Celular , Proliferación Celular , ADN/biosíntesis , Fase G1 , Hiperplasia , Masculino , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , FN-kappa B/antagonistas & inhibidores , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteína de Retinoblastoma/metabolismo
7.
Endocr J ; 56(7): 905-10, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19461158

RESUMEN

Congenital nephrogenic diabetes insipidus (CNDI) is a rare inherited disease, characterized by an inability of the kidney to concentrate urine in response to vasopressin. Three different inheritance patterns have been described, i.e., the X-linked recessive form associated with arginine vasopressin V2 receptor (AVPR2) gene mutations, the autosomal recessive and dominant forms of CNDI associated with mutations in the aquaporin-2 (AQP2) gene encoding the vasopressin-regulated water channel of the renal collecting duct. Our case is an 18-year-old male patient who complained of severe polyuria since his infancy. But his developmental and growth status were normal. He was diagnosed as CNDI by water deprivation test and genomic DNA sequencing, which revealed high plasma AVP levels but persistently low urine osmolalities to 6 h-water deprivation and the novel missense mutation S216F in exon4 of the AQP2 gene. Immunohistochemistry of renal biopsied tissue revealed that most of the AQP2 labeling was seen intracellularly in a dotted pattern in the collecting duct principal cells. Immunoblotting of urine samples revealed significantly decreased urinary excretion of AQP2 (approximately 7% of normal control). Here, we report a new case of CNDI associated with the novel missense mutation of the AQP2 gene.


Asunto(s)
Acuaporina 2/genética , Diabetes Insípida Nefrogénica/genética , Adolescente , Acuaporina 2/orina , Arginina Vasopresina/orina , Secuencia de Bases , Humanos , Masculino , Datos de Secuencia Molecular , Mutación Missense
8.
Cancer Res ; 67(6): 2595-602, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363578

RESUMEN

The cyclopentenone 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) inhibits proliferation of cancer cells, including breast cancers, by peroxisome proliferator-activated receptor-gamma (PPARgamma)-dependent and PPARgamma-independent mechanisms. However, little is known about its effect on the transcriptional activity of estrogen receptor-alpha (ERalpha) that plays vital roles in the growth of breast cancers. Here, we show that 15d-PGJ(2) inhibits both 17beta-estradiol (E(2))-dependent and E(2)-independent ERalpha transcriptional activity by PPARgamma-independent mechanism. In addition, 15d-PGJ(2) directly modifies ERalpha protein via its reactive cyclopentenone moiety, evidenced by incorporation of biotinylated 15d-PGJ(2) into ERalpha, both in vitro and in vivo. Nanoflow reverse-phase liquid chromatography tandem mass spectrometry analysis identifies two cysteines (Cys(227) and Cys(240)) within the COOH-terminal zinc finger of ERalpha DNA-binding domain (DBD) as targets for covalent modification by 15d-PGJ(2). Gel mobility shift and chromatin immunoprecipitation assays show that 15d-PGJ(2) inhibits DNA binding of ERalpha and subsequent repression of ERalpha target gene expression, such as pS2 and c-Myc. Therefore, our results suggest that 15d-PGJ(2) can block ERalpha function by covalent modification of cysteine residues within the vulnerable COOH-terminal zinc finger of ERalpha DBD, resulting in fundamental inhibition of both hormone-dependent and hormone-independent ERalpha transcriptional activity.


Asunto(s)
Receptor alfa de Estrógeno/antagonistas & inhibidores , Prostaglandina D2/análogos & derivados , Secuencia de Aminoácidos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Cisteína/metabolismo , ADN de Neoplasias/metabolismo , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Humanos , Datos de Secuencia Molecular , PPAR gamma , Prostaglandina D2/farmacología , Estructura Terciaria de Proteína , Activación Transcripcional/efectos de los fármacos , Transfección , Dedos de Zinc
9.
3 Biotech ; 8(12): 487, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30467532

RESUMEN

The objective of this study was to compare and evaluate the efficacy of collagen-based sponge compared to commercial collagen sponge as a potent open wound-dressing material. In this study, 10 mm diameter skin incision was made on lateral side of rats. The wound was monitored regularly until day 12. Histopathology results revealed the faster re-epithelialization and lesser inflammatory cells, and also masson's trichrome staining showed that collagen fibrils were horizontal and interwoven in collagen-based sponge group. The expression of growth factors such as VEGF and TGF-ß1 was found to be upregulated in transcriptional and translational levels, suggesting the importance of collagen-based sponge as a potent wound-healing material. Furthermore, IL-6 and TNF-α in the wound tissue were significantly down-regulated in 2 and 6 days in collagen-based sponge group and anti-inflammatory cytokine IL-10 level was found to be upregulated throughout 12 days. These results cumulatively revealed that collagen-based sponge may serve as novel material for wound healing in the animal model.

10.
Mol Cell Biol ; 22(14): 5203-11, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12077347

RESUMEN

Human activating signal cointegrator 1 (hASC-1) was originally isolated as a transcriptional coactivator of nuclear receptors. Here we report that ASC-1 exists as a steady-state complex associated with three polypeptides, P200, P100, and P50, in HeLa nuclei; stimulates transactivation by serum response factor (SRF), activating protein 1 (AP-1), and nuclear factor kappaB (NF-kappaB) through direct binding to SRF, c-Jun, p50, and p65; and relieves the previously described transrepression between nuclear receptors and either AP-1 or NF-kappaB. Interestingly, ectopic expression of Caenorhabditis elegans ASC-1 (ceASC-1), an ASC-1 homologue that binds P200 and P100, like hASC-1, while weakly interacting only with p65, in HeLa cells appears to replace endogenous hASC-1 from the hASC-1 complex and exerts potent dominant-negative effects on AP-1, NF-kappaB, and SRF transactivation. In addition, neutralization of endogenous P50 by single-cell microinjection of a P50 antibody inhibits AP-1 transactivation; the inhibition is relieved by coexpression of wild-type P50, but not of P50DeltaKH, a mutant form that does not interact with P200. Overall, these results suggest that the endogenous hASC-1 complex appears to play an essential role in AP-1, SRF, and NF-kappaB transactivation and to mediate the transrepression between nuclear receptors and either AP-1 or NF-kappaB in vivo.


Asunto(s)
Factores de Transcripción/química , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Núcleo Celular/metabolismo , Células HeLa , Humanos , Sustancias Macromoleculares , Datos de Secuencia Molecular , Mutación , FN-kappa B/metabolismo , Pruebas de Neutralización , Fenotipo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor de Respuesta Sérica/metabolismo , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/genética , Activación Transcripcional
11.
Nucleic Acids Res ; 31(23): 6860-72, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14627819

RESUMEN

The orphan nuclear receptor small heterodimer partner (SHP; NR0B2) is an unusual orphan nuclear receptor that lacks a conventional DNA-binding domain and acts as a modulator of transcriptional activities of a number of nuclear receptors. We have previously reported that the orphan nuclear receptor ERRgamma activates the SHP promoter. In this study, we have found that basic helix-loop-helix (bHLH) transcription factors, the E2A proteins (E47, E12 and E2/5), activated the human but not the mouse SHP promoter. In contrast, the tissue-specific E47 heterodimer partner BETA2 repressed the E47- mediated transactivation of the human SHP (hSHP) promoter. Using serial deletions and E-box mutant constructs of the hSHP promoter, we identified two E-boxes (E6 and E7) as E47-responsive E-boxes, which are not conserved in the mouse SHP promoter. Moreover, gel shift, chromatin immunoprecipitation (ChIP) and northern blot assays demonstrated that E47 directly binds to the hSHP promoter in vivo and in vitro and that Id proteins inhibited E47 binding to the hSHP promoter. Finally, we found that E47 and steroidogenic factor 1 (SF-1), a regulator of the SHP promoter, synergistically activate the human but not the mouse SHP promoter. Our findings suggest that the E2A proteins differentially regulate the human and mouse SHP promoters and cooperate with orphan nuclear receptor SF-1 for transcriptional activation of the hSHP promoter.


Asunto(s)
Regiones Promotoras Genéticas/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Activación Transcripcional , Animales , Secuencia de Bases , Línea Celular , Secuencias Hélice-Asa-Hélice , Humanos , Ratones , Unión Proteica , Proteínas Represoras/metabolismo , Elementos de Respuesta/genética , Eliminación de Secuencia/genética , Especificidad de la Especie , Factor Esteroidogénico 1
12.
Mol Endocrinol ; 18(2): 312-25, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14645497

RESUMEN

The estrogen receptor-related receptor gamma (ERR gamma/ERR3/NR3B3) is the newest member of the ERR subfamily that also includes ERR alpha and ERR beta. All three isoforms share a high degree of amino acid identity especially in the DNA binding domain. ERR gamma is a constitutively active transcriptional activator that regulates reporter elements driven by steroidogenic factor 1 response element (SF-1RE) and estrogen response element. However, it has the highest potency on a derivative of SF-1RE present in the small heterodimer partner gene promoter called sft4 and unlike ERR alpha and -beta, it fails to activate a palindromic thyroid hormone response element. To investigate the mechanism behind this response element-specific differential transcriptional activity of ERR gamma, the interactions of ERR gamma and the aforementioned response elements was monitored. EMSA and chromatin immunoprecipitation assays demonstrated that ERR gamma binds to sft4, SF-1RE, and palindromic thyroid hormone response element albeit with different degrees of affinity, but causes hyperacetylation of sft4 and SF-1RE templates only. Limited proteolysis assays showed that ERR gamma, bound to different elements, shows differential trypsin sensitivity. A search for novel coregulators of ERR gamma led to the identification of receptor interacting protein 140 as a potent corepressor and peroxisome proliferator-activated receptor gamma coactivator 1 as a potent coactivator of ERR gamma. DNA-dependent pull-down and transient transfection assays demonstrated that, on different DNA elements, ERR gamma exhibits differential cofactor interactions, which in turn dictate its transcriptional activity. Because ERR gamma shows a similar tissue distribution as peroxisome proliferator-activated receptor gamma coactivator 1 and receptor interacting protein 140, these two coregulators may act as key components of ERR gamma-mediated transcription.


Asunto(s)
ADN/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Estrógenos/genética , Elementos de Respuesta/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Células Cultivadas , ADN/metabolismo , ADN/farmacología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína de Interacción con Receptores Nucleares 1 , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Estrógenos/metabolismo , Factor Esteroidogénico 1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Activación Transcripcional , Tripsina/metabolismo
13.
Mol Endocrinol ; 18(12): 2880-94, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15358835

RESUMEN

Small heterodimer partner (SHP; NR0B2) is an atypical orphan nuclear receptor and acts as a coregulator of various nuclear receptors. Herein, we examined a novel cross talk between SHP and a forkhead transcription factor HNF3 (hepatocyte nuclear factor 3/Foxa. Transient transfection assay demonstrated that SHP inhibited the transcriptional activity of all three isoforms of HNF3, HNF3alpha, beta, and gamma. In vivo and in vitro protein interaction studies showed that SHP physically interacted with HNF3. Adenovirus-mediated overexpression of SHP significantly decreased the mRNA levels of glucose-6-phosphase (G6Pase), cholesterol 7-alpha-hydroxylase (CYP7A1), and phosphoenolpyruvate carboxykinase (PEPCK) in HepG2 cells and rat primary hepatocytes. Moreover, the mRNA level of G6Pase was notably increased by down-regulation of SHP with small interfering RNA. Interestingly, HNF3 transactivity was still repressed by SHPDelta128-139 that fails to repress nuclear receptors. Mapping of interaction domain revealed that SHP interacted with forkhead DNA binding domain of HNF3alpha. Gel mobility shift and chromatin immunoprecipitation assays demonstrated that SHP inhibits DNA binding of HNF3. These results suggest that SHP is involved in the regulation of G6Pase, CYP7A1, and PEPCK gene expression via novel mechanism of inhibition of HNF3 activity and expand the role of SHP as a coregulator of other family of transcription factors in addition to nuclear receptors.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/fisiología , Proteínas Represoras/fisiología , Factores de Transcripción/antagonistas & inhibidores , Unión Competitiva , Línea Celular , Núcleo Celular/química , Colesterol 7-alfa-Hidroxilasa/genética , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Ensayo de Cambio de Movilidad Electroforética , Glucosa-6-Fosfatasa/genética , Secuencias Hélice-Asa-Hélice/genética , Factor Nuclear 3-alfa del Hepatocito , Factor Nuclear 3-beta del Hepatocito , Factor Nuclear 3-gamma del Hepatocito , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Regiones Promotoras Genéticas/genética , Mapeo de Interacción de Proteínas , ARN Interferente Pequeño/genética , Receptores Citoplasmáticos y Nucleares/análisis , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética
14.
Mol Endocrinol ; 18(4): 776-90, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14752053

RESUMEN

Small heterodimer partner (SHP; NR0B2) is an atypical orphan nuclear receptor that lacks a conventional DNA binding domain (DBD) and represses the transcriptional activity of various nuclear receptors. In this study, we examined the novel cross talk between SHP and BETA2/NeuroD, a basic helix-loop-helix transcription factor. In vitro and in vivo protein interaction studies showed that SHP physically interacts with BETA2/NeuroD, but not its heterodimer partner E47. Moreover, confocal microscopic study and immunostaining results demonstrated that SHP colocalized with BETA2 in islets of mouse pancreas. SHP inhibited BETA2/NeuroD-dependent transactivation of an E-box reporter, whereas SHP was unable to repress the E47-mediated transactivation and the E-box mutant reporter activity. In addition, SHP repressed the BETA2-dependent activity of glucokinase and cyclin-dependent kinase inhibitor p21 gene promoters. Gel shift and in vitro protein competition assays indicated that SHP inhibits neither dimerization nor DNA binding of BETA2 and E47. Rather, SHP directly repressed BETA2 transcriptional activity and p300-enhanced BETA2/NeuroD transcriptional activity by inhibiting interaction between BETA2 and coactivator p300. We also showed that C-terminal repression domain within SHP is also required for BETA2 repression. However, inhibition of BETA2 activity was not observed by naturally occurring human SHP mutants that cannot interact with BETA2/NeuroD. Taken together, these results suggest that SHP acts as a novel corepressor for basic helix-loop-helix transcription factor BETA2/NeuroD by competing with coactivator p300 for binding to BETA2/NeuroD and by its direct transcriptional repression function.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transactivadores/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células COS , Dimerización , Proteína p300 Asociada a E1A , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Mutación , Proteínas Nucleares/metabolismo , Estructura Terciaria de Proteína , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción TCF , Proteína 1 Similar al Factor de Transcripción 7 , Factores de Transcripción/metabolismo , Activación Transcripcional
15.
Mol Endocrinol ; 18(5): 1082-95, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-14963109

RESUMEN

The orphan nuclear receptors small heterodimer partner (SHP) and dosage-sensitive sex-reversal adrenal hypoplasia congenital (AHC) critical region on the X chromosome gene 1 (DAX-1) contain extra amino acids between helices H6 and H7 of LBD, and here we investigated a possible role of these additional amino acids. Transient transfection assay demonstrated that, in contrast to wild type, in mutant SHP Delta128-139 deletion of 12 extra amino acids in H6-H7 failed to repress the transactivity of orphan nuclear receptors such as estrogen receptor-related receptor-gamma, hepatocyte nuclear factor 4alpha, and constitutive androstane receptor. Interestingly, yeast two-hybrid and glutathione-S-transferase pull-down assays demonstrated that wild-type and SHP Delta128-139 have similar abilities to interact with estrogen receptor-related receptor-gamma, hepatocyte nuclear factor 4alpha, and constitutive androstane receptor. Unexpectedly, in wild-type DAX-1 and mutant DAX-1 Delta338-362, deletion of 25 extra amino acids in H6-H7 had no significant difference in the interaction and repression of steroidogenic factor 1 transactivation. Mutant SHP that contains DAX-1 extra amino acids or polyalanine stretch in H6-H7 showed indistinguishable pattern of repression from wild-type SHP. Interestingly, the swapped SHP mutant with DAX-1 extra amino acids interacted with EID-1 (E1A-like inhibitor of differentiation 1), which is characterized as an SHP-interacting corepressor. However, interaction between SHP Delta128-139 and EID-1 was significantly diminished. Moreover, SHP-mediated repression of constitutive androstane receptor transactivation was significantly released by down-regulation of EID-1 expression with EID-1 small interfering RNA. The present study suggests that H6-H7 loop regions of SHP and DAX-1 play a different role in the repression of nuclear receptor transactivation.


Asunto(s)
Proteínas de Unión al ADN/genética , Modelos Moleculares , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Ácido Retinoico/genética , Proteínas Represoras/genética , Cromosoma X/genética , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Receptor Nuclear Huérfano DAX-1 , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Mutación/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Ácido Retinoico/metabolismo , Proteínas Represoras/metabolismo , Factor Esteroidogénico 1 , Transactivadores/genética , Transcripción Genética , Células Tumorales Cultivadas , Cromosoma X/metabolismo
16.
Sci Rep ; 5: 16577, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26560812

RESUMEN

Vascular calcification, a pathologic response to defective calcium and phosphate homeostasis, is strongly associated with cardiovascular mortality and morbidity. In this study, we have observed that pyruvate dehydrogenase kinase 4 (PDK4) is upregulated and pyruvate dehydrogenase complex phosphorylation is increased in calcifying vascular smooth muscle cells (VSMCs) and in calcified vessels of patients with atherosclerosis, suggesting that PDK4 plays an important role in vascular calcification. Both genetic and pharmacological inhibition of PDK4 ameliorated the calcification in phosphate-treated VSMCs and aortic rings and in vitamin D3-treated mice. PDK4 augmented the osteogenic differentiation of VSMCs by phosphorylating SMAD1/5/8 via direct interaction, which enhances BMP2 signaling. Furthermore, increased expression of PDK4 in phosphate-treated VSMCs induced mitochondrial dysfunction followed by apoptosis. Taken together, our results show that upregulation of PDK4 promotes vascular calcification by increasing osteogenic markers with no adverse effect on bone formation, demonstrating that PDK4 is a therapeutic target for vascular calcification.


Asunto(s)
Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Calcificación Vascular/metabolismo , Animales , Apoptosis/genética , Biomarcadores , Remodelación Ósea/genética , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Osteogénesis/genética , Fosforilación , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/química
17.
Redox Biol ; 2: 855-64, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25009787

RESUMEN

Excessive proliferation of vascular smooth muscle cells (VSMCs) and incomplete re-endothelialization is a major clinical problem limiting the long-term efficacy of percutaneous coronary angioplasty. We tested if dimethylfumarate (DMF), an anti-psoriasis drug, could inhibit abnormal vascular remodeling via NF-E2-related factor 2 (Nrf2)-NAD(P)H quinone oxidoreductase 1 (NQO1) activity. DMF significantly attenuated neointimal hyperplasia induced by balloon injury in rat carotid arteries via suppression of the G1 to S phase transition resulting from induction of p21 protein in VSMCs. Initially, DMF increased p21 protein stability through an enhancement in Nrf2 activity without an increase in p21 mRNA. Later on, DMF stimulated p21 mRNA expression through a process dependent on p53 activity. However, heme oxygenase-1 (HO-1) or NQO1 activity, well-known target genes induced by Nrf2, were dispensable for the DMF induction of p21 protein and the effect on the VSMC proliferation. Likewise, DMF protected endothelial cells from TNF-α-induced apoptosis and the dysfunction characterized by decreased eNOS expression. With knock-down of Nrf2 or NQO1, DMF failed to prevent TNF-α-induced cell apoptosis and decreased eNOS expression. Also, CD31 expression, an endothelial specific marker, was restored in vivo by DMF. In conclusion, DMF prevented abnormal proliferation in VSMCs by G1 cell cycle arrest via p21 upregulation driven by Nrf2 and p53 activity, and had a beneficial effect on TNF-α-induced apoptosis and dysfunction in endothelial cells through Nrf2-NQO1 activity suggesting that DMF might be a therapeutic drug for patients with vascular disease.


Asunto(s)
Apoptosis/efectos de los fármacos , Fumaratos/farmacología , Inmunosupresores/farmacología , Músculo Liso Vascular/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Reestenosis Coronaria/patología , Reestenosis Coronaria/prevención & control , Dimetilfumarato , Fumaratos/uso terapéutico , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Humanos , Hiperplasia/prevención & control , Inmunosupresores/uso terapéutico , Masculino , Músculo Liso Vascular/citología , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/genética , Ratas , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos
18.
PLoS One ; 8(11): e80391, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260381

RESUMEN

Scoparone, a natural compound isolated from Artemisia capillaris, has been used in Chinese herbal medicine to treat neonatal jaundice. Signal transducer and activator of transcription 3 (STAT3) contributes to the growth and survival of many human tumors. This study was undertaken to investigate the anti-tumor activity of scoparone against DU145 prostate cancer cells and to determine whether its effects are mediated by inhibition of STAT3 activity. Scoparone inhibited proliferation of DU145 cells via cell cycle arrest in G1 phase. Transient transfection assays showed that scoparone repressed both constitutive and IL-6-induced transcriptional activity of STAT3. Western blot and quantitative real-time PCR analyses demonstrated that scoparone suppressed the transcription of STAT3 target genes such as cyclin D1, c-Myc, survivin, Bcl-2, and Socs3. Consistent with this, scoparone decreased phosphorylation and nuclear accumulation of STAT3, but did not reduce phosphorylation of janus kinase 2 (JAK2) or Src, the major upstream kinases responsible for STAT3 activation. Moreover, transcriptional activity of a constitutively active mutant of STAT3 (STAT3C) was inhibited by scoparone, but not by AG490, a JAK2 inhibitor. Furthermore, scoparone treatment suppressed anchorage-independent growth in soft agar and tumor growth of DU145 xenografts in nude mice, concomitant with a reduction in STAT3 phosphorylation. Computational modeling suggested that scoparone might bind the SH2 domain of STAT3. Our findings suggest that scoparone elicits an anti-tumor effect against DU145 prostate cancer cells in part through inhibition of STAT3 activity.


Asunto(s)
Antineoplásicos/farmacología , Cumarinas/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Artemisia/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/metabolismo , Fase G1/efectos de los fármacos , Células HCT116 , Células HT29 , Células HeLa , Células Hep G2 , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Interleucina-6/metabolismo , Janus Quinasa 2/metabolismo , Células MCF-7 , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Survivin , Transcripción Genética/efectos de los fármacos
19.
Exp Diabetes Res ; 2012: 639762, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21977023

RESUMEN

The endoplasmic reticulum (ER) is a cellular organelle responsible for multiple important cellular functions including the biosynthesis and folding of newly synthesized proteins destined for secretion, such as insulin. The ER participates in all branches of metabolism, linking nutrient sensing to cellular signaling. Many pathological and physiological factors perturb ER function and induce ER stress. ER stress triggers an adaptive signaling cascade, called the unfolded protein response (UPR), to relieve the stress. The failure of the UPR to resolve ER stress leads to pathological conditions such as ß-cell dysfunction and death, and type II diabetes. However, much less is known about the fine details of the control and regulation of the ER response to hyperglycemia (glucotoxicity), hyperlipidemia (lipotoxicity), and the combination of both (glucolipotoxicity). This paper considers recent insights into how the response is regulated, which may provide clues into the mechanism of ER stress-mediated ß-cell dysfunction and death during the progression of glucolipotoxicity-induced type II diabetes.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Retículo Endoplásmico/metabolismo , Células Secretoras de Insulina/metabolismo , Metabolismo de los Lípidos , Animales , Apoptosis , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/patología , Humanos , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Metabolismo de los Lípidos/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Fisiológico , Respuesta de Proteína Desplegada
20.
Free Radic Biol Med ; 52(3): 671-682, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22155056

RESUMEN

Sulforaphane (SFN) is a dietary isothiocyanate that exerts chemopreventive effects via NF-E2-related factor 2 (Nrf2)-mediated induction of antioxidant/phase II enzymes, such as heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1). This work was undertaken to evaluate the effects of SFN on hepatic fibrosis and profibrotic transforming growth factor (TGF)-ß/Smad signaling, which are closely associated with oxidative stress. SFN suppressed TGF-ß-enhanced expression of α-smooth muscle actin (α-SMA), a marker of hepatic stellate cell (HSC) activation, and profibrogenic genes such as type I collagen, fibronectin, tissue inhibitor of matrix metalloproteinase (TIMP)-1, and plasminogen activator inhibitor (PAI)-1 in hTERT, an immortalized human HSC line. SFN inhibited TGF-ß-stimulated activity of a PAI-1 promoter construct and (CAGA)(9) MLP-Luc, an artificial Smad3/4-specific reporter, in addition to reducing phosphorylation and nuclear translocation of Smad3. Nrf2 overexpression was sufficient to inhibit the TGF-ß/Smad signaling and PAI-1 expression. Conversely, knockdown of Nrf2, but not inhibition of HO-1 or NQO1 activity, significantly abolished the inhibitory effect of SFN on (CAGA)(9) MLP-Luc activity. However, inhibition of NQO1 activity reversed repression of TGF-ß-stimulated expression of type I collagen by SFN, suggesting the involvement of antioxidant activity of SFN in the suppression of Smad-independent fibrogenic gene expression. Finally, SFN treatment attenuated the development and progression of early stage hepatic fibrosis induced by bile duct ligation in mice, accompanied by reduced expression of type I collagen and α-SMA. Collectively, these results show that SFN elicits an antifibrotic effect on hepatic fibrosis through Nrf2-mediated inhibition of the TGF-ß/Smad signaling and subsequent suppression of HSC activation and fibrogenic gene expression.


Asunto(s)
Hepatopatías/prevención & control , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Smad/metabolismo , Tiocianatos/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Inducción Enzimática/efectos de los fármacos , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibrosis , Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Isotiocianatos , Hepatopatías/patología , Luciferasas/biosíntesis , Luciferasas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Serpina E2/genética , Serpina E2/metabolismo , Transducción de Señal , Proteínas Smad/antagonistas & inhibidores , Sulfóxidos , Tiocianatos/uso terapéutico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA